25 results on '"Brian Gudenas"'
Search Results
2. Depletion of kinesin motor KIF20A to target cell fate control suppresses medulloblastoma tumour growth
- Author
-
Runxiang Qiu, Jun Wu, Brian Gudenas, Paul A. Northcott, Robert J. Wechsler-Reya, and Qiang Lu
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Runxiang Qiu et al find that conditional knockout of Kif20a, a regulator of cytokinesis and neural progenitor cell fate, induces early cell cycle exit and precocious neuronal differentiation of cerebellar granule neuron progenitors. They show that Kif20a depletion suppresses tumour formation in genetic and xenograft mouse models of medulloblastoma, indicating the value of targeting daughter cell fate specification.
- Published
- 2021
- Full Text
- View/download PDF
3. Molecular classification and outcome of children with rare CNS embryonal tumors: results from St. Jude Children’s Research Hospital including the multi-center SJYC07 and SJMB03 clinical trials
- Author
-
Anthony P. Y. Liu, Sandeep K. Dhanda, Tong Lin, Edgar Sioson, Aksana Vasilyeva, Brian Gudenas, Ruth G. Tatevossian, Sujuan Jia, Geoffrey Neale, Daniel C. Bowers, Tim Hassall, Sonia Partap, John R. Crawford, Murali Chintagumpala, Eric Bouffet, Geoff McCowage, Alberto Broniscer, Ibrahim Qaddoumi, Greg Armstrong, Karen D. Wright, Santhosh A. Upadhyaya, Anna Vinitsky, Christopher L. Tinkle, John Lucas, Jason Chiang, Daniel J. Indelicato, Robert Sanders, Paul Klimo, Frederick A. Boop, Thomas E. Merchant, David W. Ellison, Paul A. Northcott, Brent A. Orr, Xin Zhou, Arzu Onar-Thomas, Amar Gajjar, and Giles W. Robinson
- Subjects
Central Nervous System Neoplasms ,Cellular and Molecular Neuroscience ,Brain Neoplasms ,Humans ,Neuroectodermal Tumors, Primitive ,Forkhead Transcription Factors ,Neurology (clinical) ,Neoplasms, Germ Cell and Embryonal ,Child ,Glioblastoma ,Hospitals ,Pathology and Forensic Medicine - Abstract
Methylation profiling has radically transformed our understanding of tumors previously called central nervous system primitive neuro-ectodermal tumors (CNS-PNET). While this marks a momentous step toward defining key differences, reclassification has thrown treatment into disarray. To shed light on response to therapy and guide clinical decision-making, we report outcomes and molecular features of children with CNS-PNETs from two multi-center risk-adapted studies (SJMB03 for patients ≥ 3 years; SJYC07 for patients 3 years) complemented by a non-protocol institutional cohort. Seventy patients who had a histological diagnosis of CNS-PNET or CNS embryonal tumor from one of the new categories that has supplanted CNS-PNET were included. This cohort was molecularly characterized by DNA methylation profiling (n = 70), whole-exome sequencing (n = 53), RNA sequencing (n = 20), and germline sequencing (n = 28). Clinical characteristics were detailed, and treatment was divided into craniospinal irradiation (CSI)-containing (SJMB03 and SJMB03-like) and CSI-sparing therapy (SJYC07 and SJYC07-like). When the cohort was analyzed in its entirety, no differences were observed in the 5-year survival rates even when CSI-containing therapy was compared to CSI-sparing therapy. However, when analyzed by DNA methylation molecular grouping, significant survival differences were observed, and treatment particulars provided suggestions of therapeutic response. Patients with CNS neuroblastoma with FOXR2 activation (CNS-NB-FOXR2) had a 5-year event-free survival (EFS)/overall survival (OS) of 66.7% ± 19.2%/83.3% ± 15.2%, and CIC rearranged sarcoma (CNS-SARC-CIC) had a 5-year EFS/OS both of 57.1% ± 18.7% with most receiving regimens that contained radiation (focal or CSI) and multidrug chemotherapy. Patients with high-grade neuroepithelial tumor with BCOR alteration (HGNET-BCOR) had abysmal responses to upfront chemotherapy-only regimens (5-year EFS = 0%), but survival extended with salvage radiation after progression [5-year OS = 53.6% ± 20.1%]. Patients with embryonal tumor with multilayered rosettes (ETMR) or high-grade glioma/glioblastoma multiforme (HGG/GBM) did not respond favorably to any modality (5-year EFS/OS = 10.7 ± 5.8%/17.9 ± 7.2%, and 10% ± 9.0%/10% ± 9.0%, respectively). As an accompaniment, we have assembled this data onto an interactive website to allow users to probe and query the cases. By reporting on a carefully matched clinical and molecular cohort, we provide the needed insight for future clinical management.
- Published
- 2022
- Full Text
- View/download PDF
4. Depletion of kinesin motor KIF20A to target cell fate control suppresses medulloblastoma tumour growth
- Author
-
Robert J. Wechsler-Reya, Brian Gudenas, Runxiang Qiu, Paul A. Northcott, Jun Wu, and Qiang Lu
- Subjects
0301 basic medicine ,Cell division ,QH301-705.5 ,Medicine (miscellaneous) ,Kinesins ,Cell fate determination ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neural Stem Cells ,Cancer stem cell ,Animals ,Hedgehog Proteins ,Progenitor cell ,Biology (General) ,Cerebellar Neoplasms ,Mitosis ,Cell Proliferation ,Mice, Knockout ,Neurons ,Cancer stem cells ,Stem Cells ,Cell Cycle ,Cell Differentiation ,Cell cycle ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cell Fate Control ,General Agricultural and Biological Sciences ,Cytokinesis ,Medulloblastoma ,Signal Transduction - Abstract
During mammalian brain development, neural progenitor cells proliferate extensively but can ensure the production of correct numbers of various types of mature cells by balancing symmetric proliferative versus asymmetric differentiative cell divisions. This process of cell fate determination may be harnessed for developing cancer therapy. Here, we test this idea by targeting KIF20A, a mitotic kinesin crucial for the control of cell division modes, in a genetic model of medulloblastoma (MB) and human MB cells. Inducible Kif20a knockout in both normal and MB-initiating granule neuron progenitors (GNPs) causes early cell cycle exit and precocious neuronal differentiation without causing cytokinesis failure and suppresses the development of Sonic Hedgehog (SHH)-activated MB. Inducible KIF20A knockdown in human MB cells inhibits proliferation both in cultures and in growing tumors. Our results indicate that targeting the fate specification process of nascent daughter cells presents a novel avenue for developing anti-proliferation treatment for malignant brain tumors., Runxiang Qiu et al find that conditional knockout of Kif20a, a regulator of cytokinesis and neural progenitor cell fate, induces early cell cycle exit and precocious neuronal differentiation of cerebellar granule neuron progenitors. They show that Kif20a depletion suppresses tumour formation in genetic and xenograft mouse models of medulloblastoma, indicating the value of targeting daughter cell fate specification.
- Published
- 2021
5. Clinical and molecular heterogeneity of pineal parenchymal tumors: a consensus study
- Author
-
Christelle Dufour, Annie Huang, Nancy Bouvier, Cynthia Hawkins, Brian Gudenas, Eric Bouffet, Matthias A. Karajannis, Marcel Kool, Alexandru Szathmari, Cécile Faure-Conter, Amar Gajjar, Stefan Rutkowski, Brent A. Orr, Jordan R. Hansford, Stefan M. Pfister, Anthony P. Y. Liu, Arzu Onar-Thomas, Eugene Hwang, Martin Mynarek, Ho Keung Ng, Elke Pfaff, Felix Sahm, Thomas E. Merchant, Alexandre Vasiljevic, Giles W. Robinson, Paul A. Northcott, Katja von Hoff, Bryan K. Li, David T.W. Jones, Matija Snuderl, Max Levine, and Marc K. Rosenblum
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Adolescent ,Age at diagnosis ,Disease ,Bioinformatics ,Pineal Gland ,Molecular heterogeneity ,Article ,Pathology and Forensic Medicine ,Cohort Studies ,Transcriptome ,Intermediate differentiation ,Young Adult ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Internal medicine ,Humans ,Medicine ,Child ,Pineoblastoma ,Brain Neoplasms ,business.industry ,Infant, Newborn ,Infant ,DNA Methylation ,Middle Aged ,Diagnostic classification ,Clinical trial ,030104 developmental biology ,Pineal Parenchymal Tumors ,Child, Preschool ,DNA methylation ,Female ,Neurology (clinical) ,business ,Pinealoma ,MicroRNA processing ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
BackgroundRecent genomic studies have shed light on the biology and inter-tumoral heterogeneity underlying pineal parenchymal tumors, in particular pineoblastomas (PBs) and pineal parenchymal tumors of intermediate differentiation (PPTIDs). Previous reports, however, had modest sample sizes and lacked power to integrate molecular and clinical findings. The different proposed subgroup structures also highlighted a need to reach consensus on a robust and relevant classification system.MethodsWe performed a meta-analysis on 221 patients with molecularly characterized PBs and PPTIDs. DNA methylation profiles were analyzed through complementary bioinformatic approaches and molecular subgrouping was harmonized. Demographic, clinical and genomic features of patients and samples from these pineal tumor subgroups were annotated.FindingsFour clinically and biologically relevant consensus PB subgroups were defined: PB-miRNA1 (n=96), PB-miRNA2 (n=23), PB-MYC/FOXR2 (n=34) and PB-RB1 (n=25); with PPTID (n=43) remaining as a molecularly distinct entity. Genomic and transcriptomic profiling allowed the characterization of oncogenic drivers for individual subgroups, specifically, alterations in the microRNA processing pathway in PB-miRNA1/2, MYC amplification and FOXR2 overexpression in PB-MYC/FOXR2, RB1 alteration in PB-RB1, and KBTBD4 insertion in PPTID. Age at diagnosis, sex predilection and metastatic status varied significantly among tumor subgroups. While patients with PB-miRNA2 and PPTID had superior outcome, survival was intermediate for patients with PB-miRNA1, and dismal for those with PB-MYC/FOXR2 and PB-RB1.InterpretationWe systematically interrogated the clinical and molecular heterogeneity within pineal parenchymal tumors and proposed a consensus nomenclature for disease subgroups, laying the groundwork for future studies as well as routine use in tumor classification.
- Published
- 2021
- Full Text
- View/download PDF
6. ETMR-14. The single-cell landscape of pineoblastoma identifies developmental origins and exposes novel therapeutic vulnerabilities
- Author
-
Brian Gudenas, Bernhard Englinger, Anthony P Y Liu, Sheikh Tanveer Ahmad, David Meredith, Elke Pfaff, Leena Paul, Jennifer Hadley, Melissa Batts, Paul Klimo, Frederick A Boop, Amar Gajjar, Giles Robinson, Brent Orr, Hong Lin, Sanda Alexandrescu, David T W Jones, Mariella G Filbin, and Paul A Northcott
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Pineoblastoma (PB) is a rare and aggressive childhood brain tumor with highly variable age and treatment-associated outcomes. Our recent bulk tumor analyses of DNA methylation and mutational landscapes uncovered four discrete PB molecular subgroups (PB-miRNA1, PB-miRNA2, PB-MYC/FOXR2, and PB-RB), providing a major advance in our understanding of biological and clinical heterogeneity. However, developmental origins of PB subgroup heterogeneity and mechanisms governing how specific genetic alterations promote malignancy remain unknown. To resolve the cellular origins of PB, we assembled a large single-nucleus RNA-sequencing cohort (n=32) of primary PB tumors, including representatives from each subgroup. Transcriptomic analysis identified subgroup-specific gene expression programs driving intra-tumoral heterogeneity. In addition, we discovered substantial differences in the expression of miRNA biogenesis genes between the PB-miRNA1 and PB-miRNA2 subgroups, providing mechanistic support for their distinct subgroup identities despite overlapping driver events. The MYC/FOXR2 subgroup was characterized by over-expression of the FOXR2 proto-oncogene in bulk RNA-seq, which we validated in single-nuclei and identified co-expressed downstream target genes. To map PB subgroups to their putative developmental beginnings, we created a single-cell transcriptional atlas of the murine pineal gland across 11 developmental stages (E11-P21). Trajectory inference within the developing pineal gland revealed a differentiation continuum of early, mid, and mature alpha-/beta pinealocytes. Cross-species correlation and deconvolution identified significant associations between multiple PB subgroups and specific differentiation states of the pinealocyte lineage, suggestive of developmental origins. Characterization of pinealocyte development informed generation of biologically faithful disease models, including a novel genetically engineered mouse model of the PB-RB subgroup. PB-Rb1 mouse tumors were histologically and molecularly validated for their fidelity to human tumor counterparts, exhibiting up-regulation of key pinealocyte lineage markers that are diagnostic in patients. Finally, high-throughput drug screening identified several promising pharmacological candidates that may attenuate consequences of Rb1 deficiency in affected children.
- Published
- 2022
- Full Text
- View/download PDF
7. Patient-derived models recapitulate heterogeneity of molecular signatures and drug response in pediatric high-grade glioma
- Author
-
Amar Gajjar, Jinghui Zhang, Yingzhe Wang, Xiaoyan Zhu, William Caufield, Chen He, Xiaoyu Li, Burgess B. Freeman, Zoran Rankovic, Gang Wu, Michelle Monje, Nathaniel R. Twarog, Duane G. Currier, Taosheng Chen, Kimberly S Mercer, Barbara Jonchere, Jason Chiang, Ke Xu, Lawryn H. Kasper, Jia Xie, Laura D. Hover, Ibrahim Qaddoumi, Christopher L. Tinkle, Cynthia Wetmore, Wenwei Lin, Anang A. Shelat, Paul Klimo, Giles W. Robinson, Santhosh A. Upadhyaya, Suzanne J. Baker, Brent A. Orr, Alberto Broniscer, Frederick A. Boop, Paige S. Dunphy, Martine F. Roussel, Paul A. Northcott, Brian Gudenas, Chang-Hyuk Kwon, James T. Dalton, and Junyuan Zhang
- Subjects
0301 basic medicine ,Science ,General Physics and Astronomy ,Context (language use) ,Disease ,General Biochemistry, Genetics and Molecular Biology ,Article ,Paediatric cancer ,03 medical and health sciences ,Genetic Heterogeneity ,Mice ,0302 clinical medicine ,In vivo ,Glioma ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cancer models ,Child ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Cancer ,Cell Proliferation ,Regulation of gene expression ,Multidisciplinary ,business.industry ,Brain Neoplasms ,TOR Serine-Threonine Kinases ,General Chemistry ,medicine.disease ,Xenograft Model Antitumor Assays ,High-Throughput Screening Assays ,CNS cancer ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,030220 oncology & carcinogenesis ,DNA methylation ,Mutation ,Cancer research ,business - Abstract
Pediatric high-grade glioma (pHGG) is a major contributor to cancer-related death in children. In vitro and in vivo disease models reflecting the intimate connection between developmental context and pathogenesis of pHGG are essential to advance understanding and identify therapeutic vulnerabilities. Here we report establishment of 21 patient-derived pHGG orthotopic xenograft (PDOX) models and eight matched cell lines from diverse groups of pHGG. These models recapitulate histopathology, DNA methylation signatures, mutations and gene expression patterns of the patient tumors from which they were derived, and include rare subgroups not well-represented by existing models. We deploy 16 new and existing cell lines for high-throughput screening (HTS). In vitro HTS results predict variable in vivo response to PI3K/mTOR and MEK pathway inhibitors. These unique new models and an online interactive data portal for exploration of associated detailed molecular characterization and HTS chemical sensitivity data provide a rich resource for pediatric brain tumor research., Patient-derived xenografts provide a resource for basic and translational cancer research. Here, the authors generate multiple pediatric high-grade glioma xenografts, use omics technologies to show that they are representative of primary tumours and use them to assess therapeutic response.
- Published
- 2021
8. WNT-activated embryonal tumors of the pineal region
- Author
-
Cinzia Lavarino, Lili-Naz Hazrati, Brian Gudenas, Loudy P. Priesterbach-Ackley, Bryan K. Li, Nagore G. Olaciregui, Giles W. Robinson, Eric Bouffet, Michael Fisher, Pieter Wesseling, Brent A. Orr, Annie Huang, Anthony P. Y. Liu, Amar Gajjar, Vicente Santa-María López, Roel E. Reddingius, Mariona Suñol, Paul A. Northcott, CCA - Cancer biology and immunology, Pathology, and Amsterdam Neuroscience - Brain Imaging
- Subjects
Male ,Adolescent ,Pineal region ,Mutation, Missense ,Biology ,Pineal Gland ,Article ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Text mining ,Humans ,Cerebellar Neoplasms ,beta Catenin ,Pineoblastoma ,Brain Neoplasms ,Extramural ,business.industry ,Wnt signaling pathway ,Neoplasms, Germ Cell and Embryonal ,Wnt Proteins ,Embryonal tumors ,Cancer research ,Female ,Neurology (clinical) ,business ,Pinealoma ,Medulloblastoma - Published
- 2020
- Full Text
- View/download PDF
9. Correction to: Risk-adapted therapy and biological heterogeneity in pineoblastoma: integrated clinico-pathological analysis from the prospective, multi-center SJMB03 and SJYC07 trials
- Author
-
Tong Lin, Sridharan Gururangan, Daniel J. Indelicato, Rahul Kumar, Frederick A. Boop, David W. Ellison, Paul A. Northcott, Brent A. Orr, Brian Gudenas, Amar Gajjar, Eric Bouffet, Thomas E. Merchant, John R. Crawford, Tim Hassall, Daniel C. Bowers, Michael Fisher, Stewart J. Kellie, Paul Klimo, Giles W. Robinson, Murali Chintagumpala, Arzu Onar-Thomas, and Anthony P. Y. Liu
- Subjects
Pineoblastoma ,Cellular and Molecular Neuroscience ,medicine.medical_specialty ,business.industry ,medicine ,Center (algebra and category theory) ,Clinico pathological ,Neurology (clinical) ,Radiology ,business ,Pathology and Forensic Medicine - Published
- 2019
- Full Text
- View/download PDF
10. MEDB-42. GermlineElp1 deficiency promotes genomic instability and survival of granule neuron progenitors primed for SHH medulloblastoma pathogenesis
- Author
-
Jesus Garcia-Lopez, Shiekh Tanveer Ahmad, Yiran Li, Brian Gudenas, Marija Kojic, Friedrik Manz, Barbara Jonchere, Anand Mayasundari, Aaron Pitre, Jennifer Hadley, Leena Paul, Melissa Batts, Brandon Bianski, Christopher Tinkle, Brent Orr, Zoran Rankovic, Giles Robinson, Martine Roussel, Brandon Wainwright, Lena Kutscher, Hong Lin, and Paul Northcott
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Germline loss-of-function (LOF) mutations in Elongator complex protein 1 (ELP1) are found in 15-20% of childhood SHH medulloblastoma (MB) and are exceedingly rare in non-SHH-MB or other cancers. ELP1 germline carriers that develop SHH-MB harbor frequent somatic PTCH1 mutations and universally sustain loss-of-heterozygosity of the remaining ELP1 allele through chromosome 9q deletion. ELP1 functions as a scaffolding subunit of the Elongator complex that is required for posttranscriptional modification of tRNAs and maintenance of efficient translational elongation and protein homeostasis. However, the molecular, biochemical, and cellular mechanisms by which ELP1/Elongator LOF contribute to SHH-MB tumorigenesis remain largely unknown. Herein, we report that mice harboring germline Elp1 monoallelic loss (i.e., Elp1+/-) exhibit hallmark features of malignant predisposition in developing cerebellar granule neuron progenitors (GNPs), the lineage-of-origin for SHH-MB. Elp1+/- GNPs are characterized by increased replication stress-induced DNA damage, upregulation of the homologous recombination repair pathway, aberrant cell cycle, and attenuation of p53-dependent apoptosis. CRISPR/Cas9-mediated Elp1 and Ptch1 gene targeting in mouse GNPs reproduces highly penetrant SHH-MB tumors recapitulating the molecular and phenotypic features of patient tumors. Reactivation of the p53 pathway through MDM2 and PAK4 inhibitors promotes selective cell death in patient-derived xenograft tumors (PDX) harboring deleterious ELP1 mutations. Together, our findings reveal that germline Elp1 deficiency heightens genomic instability and survival in GNPs, providing a mechanistic model for the subgroup-restricted pattern of predisposition and malignancy associated with pathogenic ELP1 germline carriers. These results provide rationale for further preclinical studies evaluating drugs that overcome p53 pathway inhibition in ELP1-associated SHH-MB and a renewed outlook for improving treatment options for affected children and their families.*, # Contributed equally
- Published
- 2022
- Full Text
- View/download PDF
11. A genetic mouse model with postnatal Nf1 and p53 loss recapitulates the histology and transcriptome of human malignant peripheral nerve sheath tumor
- Author
-
Joshua Paré, Paul A. Northcott, Hongjian Jin, Angela C. Hirbe, Xinwei Cao, Brian Gudenas, Michael R. Clay, Yiping Fan, Akira Inoue, and Laura J. Janke
- Subjects
Hippo signaling pathway ,Somatic cell ,cross-species comparison ,nervous system ,Malignant peripheral nerve sheath tumor ,Biology ,medicine.disease ,Germline ,Transcriptome ,Oncology ,CDKN2A ,single-sample GSEA ,Genetically Engineered Mouse ,Basic and Translational Investigations ,Knockout mouse ,medicine ,Cancer research ,AcademicSubjects/MED00300 ,YAP/TAZ ,Surgery ,AcademicSubjects/MED00310 ,Schwann cells ,Neurology (clinical) - Abstract
BackgroundMalignant peripheral nerve sheath tumors (MPNST) are aggressive sarcomas. Somatic inactivation of NF1 and cooperating tumor suppressors, including CDKN2A/B, PRC2, and p53, is found in most MPNST. Inactivation of the LATS1/2 kinases of the Hippo pathway was recently shown to cause tumors resembling MPNST histologically, although Hippo pathway mutations are rarely found in MPNST. Because existing genetically engineered mouse (GEM) models of MPNST do not recapitulate some of the key genetic features of human MPNST, we aimed to establish a mouse MPNST model that recapitulated the human disease genetically, histologically, and molecularly.MethodsWe combined two genetically modified alleles, an Nf1;Trp53 cis-conditional allele and an inducible Plp-CreER allele (NP-Plp), to model the somatic, possibly postnatal, mutational events in human MPNST. We also generated conditional Lats1;Lats2 knockout mice. We performed histopathologic analysis of mouse MPNST models and transcriptomic comparison of mouse models and human nerve sheath tumors.ResultsPostnatal Nf1;Trp53 cis-deletion resulted in GEM-MPNST that was histologically more similar to human MPNST than the widely used germline Nf1;Trp53 cis-heterozygous (NPcis) model and showed partial loss of H3K27me3. At the transcriptome level, Nf1;p53-driven GEM-MPNST were distinct from Lats-driven GEM-MPNST and resembled human MPNST more closely than do Lats-driven tumors.ConclusionsThe NP-Plp model recapitulates human MPNST genetically, histologically, and molecularly.Key PointsPostnatal Nf1;p53 cis-deletion in NP-Plp mice results in tumors similar to MPNST.The transcriptomes of Nf1;p53-driven and Lats-driven MPNST models are distinct.NP-Plp model resembles human MPNST genetically, histologically, and molecularly.Importance of the StudyMalignant peripheral nerve sheath tumors (MPNST) are aggressive sarcomas with a poor prognosis and limited treatment options. Existing genetically engineered mouse (GEM) models of MPNST do not recapitulate some of the key genetic features of human MPNST. To model the somatic, possibly postnatal, mutational events seen in MPNST patients, we generated a GEM-MPNST model by combining two genetically modified alleles, an Nf1;Trp53 cis-conditional allele and a Plp-CreER allele. Our histologic and transcriptomic analyses showed that this NP-Plp model resembles human MPNST genetically, histologically, and molecularly—more so than the widely used NPcis model and the recently published Lats-driven model. The NP-Plp model is genetically simple, making it easy to maintain and an ideal platform for preclinical studies. Given its tamoxifen-inducible nature, this model can be used to study the time/stage dependency of the tumorigenic potential of Schwann cells.
- Published
- 2021
- Full Text
- View/download PDF
12. Patient-Derived Orthotopic Xenografts and Cell Lines from Pediatric High-Grade Glioma Recapitulate the Heterogeneity of Histopathology, Molecular Signatures, and Drug Response
- Author
-
Chen He, Brian Gudenas, James Dalton, Jason Chiang, Zoran Rankovic, Chang-Hyuk Kwon, Nathaniel R. Twarog, Wenwei Lin, Amar Gajjar, Michelle Monje, Ke Xu, Lawryn H. Kasper, Laura D. Hover, Brent A. Orr, Taosheng Chen, Burgess B. Freeman, Gang Wu, Kimberly S Mercer, Santhosh A. Upadhyaya, Paul Klimo, Yingzhe Wang, Giles W. Robinson, William Caufield, Paige S. Dunphy, Martine F. Roussel, Suzanne J. Baker, Ibrahim Qaddoumi, Cynthia Wetmore, Xiaoyu Li, Jinghui Zhang, Anang A. Shelat, Duane G. Currier, Barbara Jonchere, Christopher L. Tinkle, Paul A. Northcott, Alberto Broniscer, Frederick A. Boop, Junyuan Zhang, and Xiaoyan Zhu
- Subjects
Pathogenesis ,In vivo ,Glioma ,DNA methylation ,Cancer research ,medicine ,Context (language use) ,Biology ,Signal transduction ,medicine.disease ,PI3K/AKT/mTOR pathway ,In vitro - Abstract
Pediatric high-grade glioma (pHGG) is a major contributor to cancer-related death in children.In vitroandin vivodisease models reflecting the intimate connection between developmental context and pathogenesis of pHGG are essential to advance understanding and identify therapeutic vulnerabilities. We established 21 patient-derived pHGG orthotopic xenograft (PDOX) models and eight matched cell lines from diverse groups of pHGG. These models recapitulated histopathology, DNA methylation signatures, mutations and gene expression patterns of the patient tumors from which they were derived, and included rare subgroups not well-represented by existing models. We deployed 16 new and existing cell lines for high-throughput screening (HTS).In vitroHTS results predicted variablein vivoresponse to inhibitors of PI3K/mTOR and MEK signaling pathways. These unique new models and an online interactive data portal to enable exploration of associated detailed molecular characterization and HTS chemical sensitivity data provide a rich resource for pediatric brain tumor research.
- Published
- 2020
- Full Text
- View/download PDF
13. TMOD-06. LOSS OF DICER COOPERATES WITH TUMOR SUPPRESSORS TO INITIATE METASTATIC MEDULLOBLASTOMA
- Author
-
Sheila R. Alcantara Llaguno, Olivier Saulnier, Andrea Ventura, Michael D. Taylor, Daochun Sun, Dennis K. Burns, Brian Gudenas, Luis F. Parada, Inga Nazarenko, Paul A. Northcott, Tejus Bale, Gaspare La Rocca, Alicia Pedraza, and Yuntao Chen
- Subjects
Medulloblastoma ,Cancer Research ,business.industry ,Biology ,medicine.disease ,law.invention ,Text mining ,Oncology ,Models ,law ,Cancer research ,medicine ,biology.protein ,AcademicSubjects/MED00300 ,Suppressor ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business ,Dicer - Abstract
To determine the role of microRNA regulation in brain tumor development, we incorporated a conditional allele of the microRNA processing enzyme Dicer to a previously characterized glioma mouse model based on inactivation of the tumor suppressors Nf1, Trp53, and Pten using the Nestin-creERT2 transgene. Loss of Dicer and tumor suppressors at adult ages led to glioma development; however, mutant mice tamoxifen induced at early postnatal ages developed medulloblastoma instead of glioma. The switch in tumor spectrum occurred with 100% penetrance and tumors were histologically indistinguishable from human medulloblastoma (MB). The minimum genetic mutations required for MB formation were Dicer and Trp53. Nf1 was dispensable, while additional loss of Pten produced more invasive tumors and leptomeningeal metastases. The time window for initiation of tumorigenesis was until the 2nd postnatal week, coinciding with the disappearance of the external granule layer (EGL), where cerebellar granule neuron precursors (CGNPs) undergo proliferation. Analysis of pre-symptomatic mutant mice showed proliferative defects and retained cells in the EGL, suggesting that the tumors may arise from CGNPs. However, targeting a subset of CGNPs using Math1-creERT2 did not lead to MB development, suggesting that an earlier EGL precursor may be required for tumorigenesis. Analysis of tumor transcriptome and MB subtype-specific genes and markers show that Dicer tumors most resemble extremely high risk p53-mutated SHH MB. Small RNA and mRNA sequencing analyses showed downregulation of microRNAs and dysregulation of its targets such as N-Myc. These studies demonstrate a role for microRNAs in MB development and show a fully penetrant genetic mouse model of highly metastatic MB.
- Published
- 2021
- Full Text
- View/download PDF
14. Germline Elongator mutations in sonic hedgehog medulloblastoma
- Author
-
Michael Rusch, Aksana Vasilyeva, Marc Remke, Paul A. Northcott, Tanvi Sharma, Finn Wesenberg, Andrey Korshunov, Peter Lichter, Kristian W. Pajtler, Natalie Jäger, Sonia Partap, Till Milde, John R. Crawford, Amar Gajjar, Stefan Rutkowski, Nicholas G. Gottardo, Kyle S. Smith, Daniel C. Bowers, Christoffer Johansen, Sebastian M. Waszak, Tobias Rausch, Christelle Dufour, Damarys Loew, David T.W. Jones, Geoffrey Neale, Olaf Witt, Tone Eggen, Ivo Buchhalter, Olivier Ayrault, Dominik Sturm, Maria Feychting, Jesus Garcia-Lopez, Michael A. Grotzer, Claudia E. Kuehni, Emilie Indersie, Brandon J. Wainwright, Stéphanie Puget, Joy Nakitandwe, Marcel Kool, David W. Ellison, Marina Ryzhova, Jules Kerssemakers, Birgitta Lannering, Amy A Smith, Brent A. Orr, Joachim Schüz, Tina Veje Andersen, Murali Chintagumpala, Brian Gudenas, Bérangère Lombard, Antoine Forget, Laurence Brugières, Marija Kojic, Kim E. Nichols, Jennifer Hadley, Martin Röösli, Kristina Kjærheim, Anne Bendel, Stefan M. Pfister, Kayla V. Hamilton, Ruth G. Tatevossian, Giles W. Robinson, Jan O. Korbel, Institut Curie, PSL Research University, CNRS UMR, INSERM, Orsay, France. Université Paris Sud, Université Paris- Saclay, CNRS UMR 3347, INSERM U1021, Orsay, France., and Institut Curie [Paris]
- Subjects
0301 basic medicine ,Male ,[SDV]Life Sciences [q-bio] ,Germline ,Article ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,RNA, Transfer ,Genetic predisposition ,medicine ,Humans ,Sonic hedgehog ,Cerebellar Neoplasms ,Child ,ComputingMilieux_MISCELLANEOUS ,Germ-Line Mutation ,Genetics ,Medulloblastoma ,Multidisciplinary ,biology ,Cancer ,medicine.disease ,3. Good health ,Pedigree ,030104 developmental biology ,PTCH1 ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Translational elongation ,Transcriptional Elongation Factors - Abstract
Cancer genomics has illuminated a wide spectrum of genes and core molecular processes contributing to human malignancy. Still, the genetic and molecular basis of many cancers remains only partially explained. Genetic predisposition accounts for 5-10% of cancer diagnoses(1,2) and genetic events cooperating with known somatic driver events are poorly understood. Analyzing established cancer predisposition genes in medulloblastoma (MB), a malignant childhood brain tumor, we recently identified pathogenic germline variants that account for 5% of all MB patients(3). Here, by extending our previous analysis to include all protein-coding genes, we discovered and replicated rare germline loss-of-function (LoF) variants across Elongator Complex Protein 1 (ELP1) on 9q31.3 in 15% of pediatric MB(SHH) cases, thus implicating ELP1 as the most common MB predisposition gene and increasing genetic predisposition to 40% for pediatric MB(SHH). Inheritance was verified based on parent-offspring and pedigree analysis, which identified two families with a history of pediatric MB. ELP1-associated MBs were restricted to the molecular SHHα subtype(4) and were characterized by universal biallelic inactivation of ELP1 due to somatic loss of chromosome 9q. The majority of ELP1-associated MBs exhibited co-occurring somatic PTCH1 (9q22.32) alterations, suggesting that ELP1-deficiency predisposes to tumor development in combination with constitutive activation of SHH signaling. ELP1 is an essential subunit of the evolutionary conserved Elongator complex, whose primary function is to enable efficient translational elongation through tRNA modifications at the wobble (U(34)) position(5,6). Biochemical, transcriptional, and proteomic analyses revealed that ELP1-associated MB(SHH) are characterized by a destabilized core Elongator complex, loss of Elongator-dependent tRNA wobble modifications, codon-dependent translational reprogramming, and induction of the unfolded protein response (UPR), consistent with deregulation of protein homeostasis due to Elongator-deficiency in model systems(7–9). Our findings suggest that genetic predisposition to proteome instability is a previously underappreciated determinant in the pathogenesis of pediatric brain cancer. These results provide a strong rationale for further investigating the role of protein homeostasis in other cancer types and potential opportunities for novel therapeutic interference.
- Published
- 2020
- Full Text
- View/download PDF
15. Functional loss of a noncanonical BCOR–PRC1.1 complex accelerates SHH-driven medulloblastoma formation
- Author
-
Norman Mack, Britta Statz, Andrey Korshunov, Norbert Graf, Brian Gudenas, Laura Sieber, Lena M. Kutscher, Micah D. Gearhart, Patricia Benites Goncalves da Silva, Nadja V. Batora, Olivier Ayrault, Brent A. Orr, Marcel Kool, Mikio Hoshino, Sjoerd van Rijn, Kyle S. Smith, Audrey Mercier, Mikaella Vouri, Daisuke Kawauchi, Vivian J. Bardwell, Stefan M. Pfister, Paul A. Northcott, Gudrun Fleischhack, Katja von Hoff, Jessica Clark, Konstantin Okonechnikov, Ryo Shiraishi, Hopp Children's Cancer Center Heidelberg [Heidelber, Germany] (KITZ), German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ)-Heidelberg University Hospital [Heidelberg], Signalisation, radiobiologie et cancer, and Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Carcinogenesis ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Medizin ,Polycomb-Group Proteins ,medicine.disease_cause ,Germline ,law.invention ,Mice ,0302 clinical medicine ,law ,Sonic hedgehog ,Sequence Deletion ,0303 health sciences ,Mutation ,biology ,Chemistry ,Gene Expression Regulation, Neoplastic ,Patched-1 Receptor ,Histone ,030220 oncology & carcinogenesis ,PRC1 ,brain tumor ,Research Paper ,animal structures ,mouse model ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,medulloblastoma ,03 medical and health sciences ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Genetics ,medicine ,Animals ,Humans ,Hedgehog Proteins ,PRC1.1 complex ,Cerebellar Neoplasms ,BCOR ,030304 developmental biology ,Medulloblastoma ,medicine.disease ,Repressor Proteins ,Disease Models, Animal ,PTCH1 ,cerebellar granule cells ,biology.protein ,Cancer research ,Suppressor ,Developmental Biology - Abstract
Medulloblastoma is a malignant childhood brain tumor arising from the developing cerebellum. In Sonic Hedgehog (SHH) subgroup medulloblastoma, aberrant activation of SHH signaling causes increased proliferation of granule neuron progenitors (GNPs), and predisposes these cells to tumorigenesis. A second, cooperating genetic hit is often required to push these hyperplastic cells to malignancy and confer mutation-specific characteristics associated with oncogenic signaling. Somatic loss-of-function mutations of the transcriptional corepressor BCOR are recurrent and enriched in SHH medulloblastoma. To investigate BCOR as a putative tumor suppressor, we used a genetically engineered mouse model to delete exons 9/10 of Bcor (BcorΔE9–10) in GNPs during development. This mutation leads to reduced expression of C-terminally truncated BCOR (BCORΔE9–10). While BcorΔE9–10 alone did not promote tumorigenesis or affect GNP differentiation, BcorΔE9–10 combined with loss of the SHH receptor gene Ptch1 resulted in fully penetrant medulloblastomas. In Ptch1+/−;BcorΔE9–10 tumors, the growth factor gene Igf2 was aberrantly up-regulated, and ectopic Igf2 overexpression was sufficient to drive tumorigenesis in Ptch1+/− GNPs. BCOR directly regulates Igf2, likely through the PRC1.1 complex; the repressive histone mark H2AK119Ub is decreased at the Igf2 promoter in Ptch1+/−;BcorΔE9–10 tumors. Overall, our data suggests that BCOR–PRC1.1 disruption leads to Igf2 overexpression, which transforms preneoplastic cells to malignant tumors.
- Published
- 2020
- Full Text
- View/download PDF
16. EPCO-26. INTEGRATIVE MULTI-OMICS IDENTIFIES CONVERGING DEVELOPMENTAL ORIGINS OF DISTINCT MEDULLOBLASTOMA SUBGROUPS
- Author
-
Parthiv Haldipur, Volker Hovestadt, Paul Northcott, Laure Bihannic, Zoltan Patay, Igor Y. Iskusnykh, Qingsong Gao, Kimberly Aldinger, Kathleen J. Millen, Viktor V Chizhikov, Kyle S. Smith, Giles Robinson, Brian Gudenas, Brent A. Orr, Ian A. Glass, and Mei Deng
- Subjects
Medulloblastoma ,Cancer Research ,Cell lineage ,Biology ,medicine.disease ,medicine.disease_cause ,Oncology ,medicine ,Multi omics ,Neurology (clinical) ,Epigenetics ,Stem cell ,Carcinogenesis ,Rhombic lip ,Neuroscience - Abstract
Understanding the interplay between normal development and tumorigenesis, including the identification and characterization of lineage-specific origins of MB, is a fundamental challenge in the field. Recent studies have highlighted novel associations between biologically distinct MB subgroups and diverse murine cerebellar lineages via cross-species single-cell transcriptomics. Specifically, Group 4-MB correlated with the unipolar brush cell lineage and Group 3-MB resembled Nestin+ stem cells of the early cerebellum. However, these analyses were hampered by low resolution due to the sparsity of pertinent cerebellar cell types and the cross-species nature of the approach. Herein, we profoundly expand the depth of these rare developmental populations in the murine cerebellum using a combination of lineage tracing and integrative multi-omics. Isolation and enrichment of spatially and temporally unique developmental trajectories of key rhombic lip-derived glutamatergic lineages provided an enhanced reference for mapping MB subgroups based on molecular overlap, especially for poorly defined Group 3- and Group 4-MB. Further comparisons to a novel single-cell atlas of the human fetal cerebellum, companioned with laser-capture microdissected transcriptional and epigenetic datasets, reinforced developmental insights extracted from the mouse. Characterization of compartment-specific transcriptional programs and co-expression networks identified in the human upper rhombic lip implicated convergent cellular correlates of Group 3- and Group 4-MB, suggestive of a common developmental link. Together, our results strongly implicate developmental lineages of the upper rhombic lip as the probable origins of poorly defined Group 3- and Group 4-MB. These important findings will shape future efforts to accurately model the biological heterogeneity underlying these subgroups and provide unprecedented opportunities to explore their cellular and mechanistic basis.
- Published
- 2021
- Full Text
- View/download PDF
17. MBRS-24. FUNCTIONAL CHARACTERIZATION OF IKBKAP/ELP1 AS A NOVEL SHH MEDULLOBLASTOMA PREDISPOSITION GENE
- Author
-
Kyle S. Smith, Jesus Garcia Lopez, Marija Kojic, Stefan M. Pfister, Daisuke Kawauchi, Brian Gudenas, Brandon J. Wainwright, Giles W. Robinson, Amar Gajjar, Lena M. Kutscher, Paul A. Northcott, and Jennifer Hadley
- Subjects
Medulloblastoma ,Cancer Research ,animal structures ,IKBKAP ,Biology ,medicine.disease ,Medulloblastoma (Research) ,Oncology ,medicine ,Cancer research ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Gene - Abstract
Medulloblastoma (MB), a common malignant pediatric brain tumor, comprises at least four distinct molecular entities: WNT, SHH, Group 3, and Group 4. SHH-MB is driven by aberrant activation of the Sonic hedgehog (SHH) pathway in granule neuron progenitors (GNPs) and is associated with hereditary cancer predisposition syndromes including Li Fraumeni and Gorlin. We recently identified germline loss of function (LoF) mutations affecting IKBKAP/ELP1, the primary scaffolding subunit of the Elongator complex in a subset of SHH-MB patients. Germline ELP1 mutations account for ~15% of all pediatric SHH-MBs and position ELP1 as the most prevalent hereditary predisposition gene in MB. We genetically engineered Elp1 LoF in mouse GNPs to determine Elp1 function in cerebellar development and SHH-MB. Results of both mechanistic and phenotypic experiments demonstrate that GNPs harboring Elp1 loss exhibit ribosome pausing and protein aggregation, reinforcing the critical role of Elp1 in translational elongation and protein homeostasis. Further, we generated new transgenic mouse models mimicking germline ELP1 LoF mutations observed in SHH-MB patients. Elp1+/- transgenic mice exhibit purkinje cell degeneration and an increased DNA damage response. These mice are currently being evaluated for their capacity to recapitulate ELP1-associated SHH-MB. Additional analyses carried out on SHH-MB patient-derived xenografts showed that ELP1-mutant tumor cells specifically exhibit defects in tRNA biogenesis. Therefore, the function of ELP1 as a translational regulator is severely impaired in ELP1-mutant SHH-MBs. Our ongoing molecular and functional studies will provide important insights into the most common MB predisposition gene and will lay the foundation for future preclinical studies.
- Published
- 2020
18. ETMR-06. DISSECTING THE MOLECULAR AND DEVELOPMENTAL BASIS OF PINEOBLASTOMA THROUGH GENOMICS
- Author
-
David W. Ellison, Daniel C. Bowers, Paul A. Northcott, Paul Klimo, Giles W. Robinson, Jason Chiang, Thomas E. Merchant, Laure Bihannic, David T.W. Jones, Leena Paul, Anthony P. Y. Liu, Michael Fisher, Sridharan Gururangan, Eric Bouffet, David Meredith, Rahul Kumar, John R. Crawford, Murali Chintagumpala, Tim Hassall, Amar Gajjar, Bernhard Englinger, Sanda Alexandrescu, Tong Lin, Frederick A. Boop, Daniel J. Indelicato, Yiai Tong, Mariella G. Filbin, Stewart J. Kellie, Brent A. Orr, Elke Pfaff, and Brian Gudenas
- Subjects
Pineoblastoma ,Cancer Research ,Oncology ,AcademicSubjects/MED00300 ,Genomics ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Computational biology ,ETMR and other Embryonal Tumors ,Biology - Abstract
Pineoblastoma (PB) is an aggressive embryonal brain tumor comprising 1% of pediatric CNS tumors. The clinico-molecular heterogeneity and developmental origins underlying PB are poorly understood; therefore, we have assembled a molecular cohort of histologically defined PBs (n=43) with corresponding outcome data. Methylation profiling revealed four molecularly and clinically distinct PB subgroups, including two novel entities. Mutational and transcriptional analysis identified characteristic molecular features of each subgroup, such as mutations in the miRNA processing pathway or FOXR2 proto-oncogene overexpression. Furthermore, subgroups exhibited differences in propensity for metastasis, cytogenetics, and clinical outcomes. To dissect PB developmental origins and resolve PB subgroup biology, we have employed a combination of single-cell genomics and genetically engineered mouse modeling. We created a single-cell transcriptional atlas of the developing murine pineal gland across 11 timepoints and are currently integrating these data with single nuclei RNA-seq data of human PB (n=25). Single-cell analysis of the developing pineal gland revealed three distinct populations of pinealocytes, referred to as early, mid and late pinealocytes, which segregate by developmental stage yet lie along a single developmental trajectory. Preliminary results implicate significant associations between PBs and the early pinealocyte population as well as subgroup-specific differences in intratumoral heterogeneity. Furthermore, this knowledge has informed the downstream generation of biologically faithful disease models, including a transgenic mouse model of the PB-RB subgroup. Remarkably, this model shows up-regulation of key markers of PB such as Crx, Asmt and Otx2 and substantiates early pinealocytes as the probable cell-of-origin for this PB subgroup.
- Published
- 2020
19. MBCL-21. GERMLINE ELONGATOR MUTATIONS IN SONIC HEDGEHOG MEDULLOBLASTOMA
- Author
-
Michael A. Grotzer, Marina Ryzhova, Kyle S. Smith, Olaf Witt, Ivo Buchhalter, Dominic Sturm, Paul A. Northcott, David T.W. Jones, Kayla V. Hamilton, Sonia Partap, Emilie Indersie, Daniel C. Bowers, Ruth G. Tatevossian, Anne Bendel, Laurence Brugières, Till Milde, Christelle Dufour, Stefan M. Pfister, Tobias Rausch, M Remke, Jan O. Korbel, Amy M. Smith, Tanvi Sharma, Sebastian M. Waszak, Giles W. Robinson, Stefan Rutkowski, Brent A. Orr, Natalie Jäger, Andrey Korshunov, Stéphanie Puget, Olivier Ayrault, Brandon J. Wainwright, Marcel Kool, Antoine Forget, Jennifer Hadley, Marija Kojic, Kim E. Nichols, Amar Gajjar, Damarys Loew, Peter Lichter, Garcia-Lopez Jesus, Kristian W. Pajtler, John R. Crawford, Nicholas G. Gottardo, David W. Ellison, Bérangère Lombard, Brian Gudenas, and Murali Chintagumpala
- Subjects
Medulloblastoma ,Cancer Research ,Mutation ,biology ,Cancer ,PTCH1 Gene ,medicine.disease_cause ,medicine.disease ,Germline ,Oncology ,medicine ,Cancer research ,biology.protein ,Medulloblastoma (Clinical) ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Sonic hedgehog ,Protein p53 ,Genetic pedigree - Abstract
BACKGROUND Our previous analysis of established cancer predisposition genes in medulloblastoma (MB) identified pathogenic germline variants in ~5% of all patients. Here, we extended our analysis to include all protein-coding genes. METHODS Case-control analysis performed on 795 MB patients against >118,000 cancer-free children and adults was performed to identify an association between rare germline variants and MB. RESULTS Germline loss-of-function variants of Elongator Complex Protein 1 (ELP1; 9q31.3) were strongly associated with SHH subgroup (MBSHH). ELP1-associated-MBs accounted for ~15% (29/202) of pediatric MBSHH cases and were restricted to the SHHα subtype. ELP1-associated-MBs demonstrated biallelic inactivation of ELP1 due to somatic chromosome 9q loss and most tumors exhibited co-occurring somatic PTCH1 (9q22.32) alterations. Inheritance was verified by parent-offspring sequencing (n=3) and pedigree analysis identified two families with a history of pediatric MB. ELP1-associated-MBSHH were characterized by desmoplastic/nodular histology (76%; 13/17) and demonstrated a favorable clinical outcome when compared to TP53-associated-MBSHH (5-yr OS 92% vs 20%; p-value=1.3e-6) despite both belonging to the SHHα subtype. ELP1 is a subunit of the Elongator complex, that promotes efficient translational elongation through tRNA modifications at the wobble (U34) position. Biochemical, transcriptional, and proteomic analyses revealed ELP1-associated-MBs exhibit destabilization of the core Elongator complex, loss of tRNA wobble modifications, codon-dependent translational reprogramming, and induction of the unfolded protein response. CONCLUSIONS We identified ELP1 as the most common MB predisposition gene, increasing the total genetic predisposition for pediatric MBSHH to 40%. These results mark MBSHH as an overwhelmingly genetically-predisposed disease and implicate disruption of protein homeostasis in MBSHH development.
- Published
- 2020
20. Genomic data mining for functional annotation of human long noncoding RNAs
- Author
-
Liangjiang Wang, Brian Gudenas, An-qi Wei, Jun Wang, Steven Cogill, and Shuzhen Kuang
- Subjects
0301 basic medicine ,Support Vector Machine ,Autism Spectrum Disorder ,Genomics ,Computational biology ,Review ,Biology ,ENCODE ,General Biochemistry, Genetics and Molecular Biology ,Machine Learning ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,Data Mining ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,Gene ,Genomic organization ,General Veterinary ,General Medicine ,Non-coding RNA ,Long non-coding RNA ,030104 developmental biology ,030220 oncology & carcinogenesis ,Human genome ,RNA, Long Noncoding - Abstract
Life may have begun in an RNA world, which is supported by increasing evidence of the vital role that RNAs perform in biological systems. In the human genome, most genes actually do not encode proteins; they are noncoding RNA genes. The largest class of noncoding genes is known as long noncoding RNAs (lncRNAs), which are transcripts greater in length than 200 nucleotides, but with no protein-coding capacity. While some lncRNAs have been demonstrated to be key regulators of gene expression and 3D genome organization, most lncRNAs are still uncharacterized. We thus propose several data mining and machine learning approaches for the functional annotation of human lncRNAs by leveraging the vast amount of data from genetic and genomic studies. Recent results from our studies and those of other groups indicate that genomic data mining can give insights into lncRNA functions and provide valuable information for experimental studies of candidate lncRNAs associated with human disease.
- Published
- 2019
21. ETMR-21. META-ANALYSIS OF PINEAL REGION TUMOURS DEMONSTRATES MOLECULAR SUBGROUPS WITH DISTINCT CLINICO-PATHOLOGICAL FEATURES: A CONSENSUS STUDY
- Author
-
Cynthia Hawkins, Christelle Dufour, David T.W. Jones, Paul A. Northcott, Sivan Gershanov, Elke Pfaff, Arzu Onar-Thomas, Eric Bouffet, Amar Gajjar, David W. Ellison, Sridharan Gururangan, Brent A. Orr, Christian Aichmüller, Stefan M. Pfister, Anthony P. Y. Liu, Bryan K. Li, Giles W. Robinson, Eugene Hwang, Brian Gudenas, Martin Sill, Tong Lin, Matthias A. Karajannis, Alexandre Vasiljevic, Annie Laquierre, Matija Snuderl, and Annie Huang
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Oncology ,Pineal region ,Meta-analysis ,medicine ,AcademicSubjects/MED00300 ,Clinico pathological ,AcademicSubjects/MED00310 ,Neurology (clinical) ,ETMR and other Embryonal Tumors ,Biology - Abstract
Pineoblastomas (PB) are rare, aggressive pineal gland tumours with poor global OS of 50–70% and only 15–49% OS for patients
- Published
- 2020
22. Abstract 6144: St. Jude Pediatric Brain Tumor Portal: Cloud-based resource for patient-derived orthotopic xenograft (PDOX) models of pediatric high-grade glioma, ependymoma, and CNS embryonal tumors
- Author
-
Michael A. Dyer, Brandon McMahan, Ashok K. Boddu, Clay McLeod, Frederick A. Boop, Edgar Sioson, Xiaoyan Zhu, Paige S. Dunphy, Martine F. Roussel, Swapnali Mohite, Chen He, Darrell T. Gentry, Kyle S. Smith, Jason Chiang, Irina McGuire, Paul A. Northcott, Daniel Alford, Christopher L. Tinkle, Austyn Trull, Kimberly S Mercer, Paul Klimo, Giles W. Robinson, Sarah Robinson, Nathaniel R. Twarog, Suzanne J. Baker, J. Robert Michael, Junyuan Zhang, Anthony Woodard, Brent A. Orr, Jinghui Zhang, Nedra Robison, Cynthia Williams, Keith Perry, Aksana Vasilyeva, Anang A. Shelat, Brian Gudenas, Chang-Hyuk Kwon, Kirby Birch, Amar Gajjar, Xin Zhou, Ke Xu, Lawryn H. Kasper, Laura D. Hover, Gang Wu, Ed Suh, and Jon D. Larson
- Subjects
Oncology ,Ependymoma ,Cancer Research ,medicine.medical_specialty ,Embryonal tumors ,business.industry ,Internal medicine ,medicine ,Pediatric Brain Tumor ,medicine.disease ,business ,High-Grade Glioma - Abstract
Pediatric brain tumors comprise a distinct spectrum of diseases compared to adult brain tumors and are distinguished by their unique clinical and histopathological features, developmental context, mutation burden, and genomic, epigenomic, and transcriptomic alterations. Access to in vivo models that recapitulate pediatric brain tumors has been limited and inadequate to represent these heterogeneous diseases. Here we introduce the Pediatric Brain Tumor Portal (PBTP, pbtp.stjude.cloud), an open resource to access molecular characterization, including whole-genome sequencing, whole-exome sequencing, RNA-seq, and DNA methylome profiling, of patient-derived orthotopic xenograft (PDOX) models of pediatric brain tumors. The portal will host more than 70 models, which currently include pediatric High-Grade Glioma (pHGG), Medulloblastoma, Atypical Teratoid/Rhabdoid Tumors (AT/RT), Ependymoma, and Embryonal Tumors with Multilayered Rosettes (ETMR), and reflects close to ten years of effort to generate and extensively characterize in vivo models that recapitulate primary pediatric brain tumors. PBTP is a database-driven and user-friendly platform that enables multi-omics views of PDOX and matched patient tumor and germline samples at multiple levels to help identify appropriate lines for studies of tumorigenesis or preclinical testing. To evaluate the representation of different molecular features within a disease subgroup, we implemented various visualization tools for searching and comparing somatic mutations, gene expression profiles, and methylation groups of our models and matched patient tumors. Details for each tumor including de-identified clinical data, histology, growth characteristics, DNA methylation classification, mutation status, and copy number variations are also readily available for side-by-side comparison. Furthermore, in vitro chemical sensitivity profiling is presented for selected models with matched cell lines. Users can investigate genes of interest for single nucleotide variants, small insertions and deletions, copy number variations, gene fusions, and mRNA expression in PDOX and matched patient tumors. This portal has been integrated into the St. Jude Cloud platform, through which users can explore patient and PDOX sequencing data in the context of larger cohort data sets and download raw genomics data files. PBTP provides a platform to share PDOX models with in-depth genome/epigenome-wide characterization to support advances in basic and translational research in pediatric brain tumors. *co-first, #co-corresponding Citation Format: Paige S. Dunphy, Ke Xu, Darrell T. Gentry, Chen He, Kimberly Mercer, Xiaoyan Zhu, Kyle Smith, Brian Gudenas, Sarah Robinson, Junyuan Zhang, Lawryn H. Kasper, Chang-Hyuk Kwon, Laura D. Hover, Jon D. Larson, Nathaniel Twarog, Aksana Vasilyeva, Nedra Robison, Daniel Alford, Cynthia Williams, Anthony Woodard, Xin Zhou, Edgar Sioson, J. Robert Michael, Austyn Trull, Irina McGuire, Brandon McMahan, Swapnali Mohite, Ashok Boddu, Kirby Birch, Clay McLeod, Michael A. Dyer, Paul Klimo, Frederick A. Boop, Amar Gajjar, Christopher L. Tinkle, Giles Robinson, Brent A. Orr, Jason Chiang, Paul A. Northcott, Jinghui Zhang, Keith Perry, Gang Wu, Anang A. Shelat, Ed Suh, Martine F. Roussel, Suzanne J. Baker. St. Jude Pediatric Brain Tumor Portal: Cloud-based resource for patient-derived orthotopic xenograft (PDOX) models of pediatric high-grade glioma, ependymoma, and CNS embryonal tumors [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6144.
- Published
- 2020
- Full Text
- View/download PDF
23. Integrative genomic analyses for identification and prioritization of long non-coding RNAs associated with autism
- Author
-
Anand Srivastava, Liangjiang Wang, and Brian Gudenas
- Subjects
0301 basic medicine ,Pervasive Developmental Disorders ,Autism Spectrum Disorder ,Gene Identification and Analysis ,Social Sciences ,Gene Expression ,Genetic Networks ,Biochemistry ,Transcriptome ,Post-Transcriptional Gene Regulation ,Psychology ,Genetics ,Multidisciplinary ,Genomics ,Long non-coding RNA ,Nucleic acids ,Medicine ,RNA, Long Noncoding ,Transcriptome Analysis ,Network Analysis ,Research Article ,Computer and Information Sciences ,DNA Copy Number Variations ,Sequence analysis ,Science ,Biology ,ENCODE ,behavioral disciplines and activities ,03 medical and health sciences ,mental disorders ,medicine ,Humans ,Gene Regulation ,Autistic Disorder ,Non-coding RNA ,Gene ,Biology and life sciences ,Sequence Analysis, RNA ,Gene Expression Profiling ,Neurotransmission ,Computational Biology ,Genome Analysis ,medicine.disease ,Gene expression profiling ,030104 developmental biology ,Developmental Psychology ,Long non-coding RNAs ,RNA ,Autism ,Neuroscience - Abstract
Genetic studies have identified many risk loci for autism spectrum disorder (ASD) although causal factors in the majority of cases are still unknown. Currently, known ASD risk genes are all protein-coding genes; however, the vast majority of transcripts in humans are non-coding RNAs (ncRNAs) which do not encode proteins. Recently, long non-coding RNAs (lncRNAs) were shown to be highly expressed in the human brain and crucial for normal brain development. We have constructed a computational pipeline for the integration of various genomic datasets to identify lncRNAs associated with ASD. This pipeline utilizes differential gene expression patterns in affected tissues in conjunction with gene co-expression networks in tissue-matched non-affected samples. We analyzed RNA-seq data from the cortical brain tissues from ASD cases and controls to identify lncRNAs differentially expressed in ASD. We derived a gene co-expression network from an independent human brain developmental transcriptome and detected a convergence of the differentially expressed lncRNAs and known ASD risk genes into specific co-expression modules. Co-expression network analysis facilitates the discovery of associations between previously uncharacterized lncRNAs with known ASD risk genes, affected molecular pathways and at-risk developmental time points. In addition, we show that some of these lncRNAs have a high degree of overlap with major CNVs detected in ASD genetic studies. By utilizing this integrative approach comprised of differential expression analysis in affected tissues and connectivity metrics from a developmental co-expression network, we have prioritized a set of candidate ASD-associated lncRNAs. The identification of lncRNAs as novel ASD susceptibility genes could help explain the genetic pathogenesis of ASD.
- Published
- 2017
24. A Genetic Algorithm for Finding Discriminative Functional Motifs in Long Non-coding RNAs
- Author
-
Brian Gudenas and Liangjiang Wang
- Subjects
0301 basic medicine ,Regulation of gene expression ,business.industry ,RNA ,Pattern recognition ,Computational biology ,Biology ,Subcellular localization ,Transcriptome ,03 medical and health sciences ,030104 developmental biology ,Discriminative model ,Motif (music) ,Artificial intelligence ,business - Abstract
Long non-coding RNAs (lncRNAs), each with >200 nucleotides in length, constitute a large portion of the human transcriptome. Although recent studies indicate that lncRNAs play key roles in gene regulation, development and disease, the RNA functional motifs are still poorly understood. Most of the existing algorithms for motif finding are severely limited in scalability with regards to sequence and motif size. In this study, we propose a novel genetic algorithm for discriminative motif identification capable of handling large input sequences and motif sizes by utilizing genetic operators to learn and evolve in response to the input sequences. We utilize our method on long non-coding RNA (lncRNA) transcripts as a test case to identify functional motifs associated with subcellular localization. Our methodology shows high accuracy and the ability to identify functional motifs associated with subcellular localization in lncRNAs, which recapitulates a previous experimental study.
- Published
- 2017
- Full Text
- View/download PDF
25. Gene Coexpression Networks in Human Brain Developmental Transcriptomes Implicate the Association of Long Noncoding RNAs with Intellectual Disability
- Author
-
Liangjiang Wang and Brian Gudenas
- Subjects
Regulation of gene expression ,Microarray ,Applied Mathematics ,lncRNAs ,Genomics ,Computational biology ,Biology ,Proteomics ,Bioinformatics ,Biochemistry ,Computer Science Applications ,Transcriptome ,Computational Mathematics ,lcsh:Biology (General) ,intellectual disability ,coexpression network analysis ,Copy-number variation ,Molecular Biology ,Gene ,lcsh:QH301-705.5 ,Function (biology) ,Original Research - Abstract
The advent of next-generation sequencing for genetic diagnoses of complex developmental disorders, such as intellectual disability (ID), has facilitated the identification of hundreds of predisposing genetic variants. However, there still exists a vast gap in our knowledge of causal genetic factors for ID as evidenced by low diagnostic yield of genetic screening, in which identifiable genetic causes are not found for the majority of ID cases. Most methods of genetic screening focus on protein-coding genes; however, noncoding RNAs may outnumber protein-coding genes and play important roles in brain development. Long noncoding RNAs (lncRNAs) specifically have been shown to be enriched in the brain and have diverse roles in gene regulation at the transcriptional and posttranscriptional levels. LncRNAs are a vastly uncharacterized group of noncoding genes, which could function in brain development and harbor ID-predisposing genetic variants. We analyzed lncRNAs for coexpression with known ID genes and affected biological pathways within a weighted gene coexpression network derived from RNA-sequencing data spanning human brain development. Several ID-associated gene modules were found to be enriched for lncRNAs, known ID genes, and affected biological pathways. Utilizing a list of de novo and pathogenic copy number variants detected in ID probands, we identified lncRNAs overlapping these genetic structural variants. By integrating our results, we have made a prioritized list of potential ID-associated lncRNAs based on the developing brain gene coexpression network and genetic structural variants found in ID probands.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.