169 results on '"Hamid, Bolouri"'
Search Results
2. Neural G0: a quiescent‐like state found in neuroepithelial‐derived cells and glioma
- Author
-
Samantha A O’Connor, Heather M Feldman, Sonali Arora, Pia Hoellerbauer, Chad M Toledo, Philip Corrin, Lucas Carter, Megan Kufeld, Hamid Bolouri, Ryan Basom, Jeffrey Delrow, José L McFaline‐Figueroa, Cole Trapnell, Steven M Pollard, Anoop Patel, Patrick J Paddison, and Christopher L Plaisier
- Subjects
G0 ,glioma ,neural stem cells ,quiescence ,scRNA‐seq ,Biology (General) ,QH301-705.5 ,Medicine (General) ,R5-920 - Abstract
Abstract Single‐cell RNA sequencing has emerged as a powerful tool for resolving cellular states associated with normal and maligned developmental processes. Here, we used scRNA‐seq to examine the cell cycle states of expanding human neural stem cells (hNSCs). From these data, we constructed a cell cycle classifier that identifies traditional cell cycle phases and a putative quiescent‐like state in neuroepithelial‐derived cell types during mammalian neurogenesis and in gliomas. The Neural G0 markers are enriched with quiescent NSC genes and other neurodevelopmental markers found in non‐dividing neural progenitors. Putative glioblastoma stem‐like cells were significantly enriched in the Neural G0 cell population. Neural G0 cell populations and gene expression are significantly associated with less aggressive tumors and extended patient survival for gliomas. Genetic screens to identify modulators of Neural G0 revealed that knockout of genes associated with the Hippo/Yap and p53 pathways diminished Neural G0 in vitro, resulting in faster G1 transit, down‐regulation of quiescence‐associated markers, and loss of Neural G0 gene expression. Thus, Neural G0 represents a dynamic quiescent‐like state found in neuroepithelial‐derived cells and gliomas.
- Published
- 2021
- Full Text
- View/download PDF
3. SBML Level 3: an extensible format for the exchange and reuse of biological models
- Author
-
Sarah M Keating, Dagmar Waltemath, Matthias König, Fengkai Zhang, Andreas Dräger, Claudine Chaouiya, Frank T Bergmann, Andrew Finney, Colin S Gillespie, Tomáš Helikar, Stefan Hoops, Rahuman S Malik‐Sheriff, Stuart L Moodie, Ion I Moraru, Chris J Myers, Aurélien Naldi, Brett G Olivier, Sven Sahle, James C Schaff, Lucian P Smith, Maciej J Swat, Denis Thieffry, Leandro Watanabe, Darren J Wilkinson, Michael L Blinov, Kimberly Begley, James R Faeder, Harold F Gómez, Thomas M Hamm, Yuichiro Inagaki, Wolfram Liebermeister, Allyson L Lister, Daniel Lucio, Eric Mjolsness, Carole J Proctor, Karthik Raman, Nicolas Rodriguez, Clifford A Shaffer, Bruce E Shapiro, Joerg Stelling, Neil Swainston, Naoki Tanimura, John Wagner, Martin Meier‐Schellersheim, Herbert M Sauro, Bernhard Palsson, Hamid Bolouri, Hiroaki Kitano, Akira Funahashi, Henning Hermjakob, John C Doyle, Michael Hucka, SBML Level 3 Community members, Richard R Adams, Nicholas A Allen, Bastian R Angermann, Marco Antoniotti, Gary D Bader, Jan Červený, Mélanie Courtot, Chris D Cox, Piero Dalle Pezze, Emek Demir, William S Denney, Harish Dharuri, Julien Dorier, Dirk Drasdo, Ali Ebrahim, Johannes Eichner, Johan Elf, Lukas Endler, Chris T Evelo, Christoph Flamm, Ronan MT Fleming, Martina Fröhlich, Mihai Glont, Emanuel Gonçalves, Martin Golebiewski, Hovakim Grabski, Alex Gutteridge, Damon Hachmeister, Leonard A Harris, Benjamin D Heavner, Ron Henkel, William S Hlavacek, Bin Hu, Daniel R Hyduke, Hidde de Jong, Nick Juty, Peter D Karp, Jonathan R Karr, Douglas B Kell, Roland Keller, Ilya Kiselev, Steffen Klamt, Edda Klipp, Christian Knüpfer, Fedor Kolpakov, Falko Krause, Martina Kutmon, Camille Laibe, Conor Lawless, Lu Li, Leslie M Loew, Rainer Machne, Yukiko Matsuoka, Pedro Mendes, Huaiyu Mi, Florian Mittag, Pedro T Monteiro, Kedar Nath Natarajan, Poul MF Nielsen, Tramy Nguyen, Alida Palmisano, Jean‐Baptiste Pettit, Thomas Pfau, Robert D Phair, Tomas Radivoyevitch, Johann M Rohwer, Oliver A Ruebenacker, Julio Saez‐Rodriguez, Martin Scharm, Henning Schmidt, Falk Schreiber, Michael Schubert, Roman Schulte, Stuart C Sealfon, Kieran Smallbone, Sylvain Soliman, Melanie I Stefan, Devin P Sullivan, Koichi Takahashi, Bas Teusink, David Tolnay, Ibrahim Vazirabad, Axel von Kamp, Ulrike Wittig, Clemens Wrzodek, Finja Wrzodek, Ioannis Xenarios, Anna Zhukova, and Jeremy Zucker
- Subjects
computational modeling ,file format ,interoperability ,reproducibility ,systems biology ,Biology (General) ,QH301-705.5 ,Medicine (General) ,R5-920 - Abstract
Abstract Systems biology has experienced dramatic growth in the number, size, and complexity of computational models. To reproduce simulation results and reuse models, researchers must exchange unambiguous model descriptions. We review the latest edition of the Systems Biology Markup Language (SBML), a format designed for this purpose. A community of modelers and software authors developed SBML Level 3 over the past decade. Its modular form consists of a core suited to representing reaction‐based models and packages that extend the core with features suited to other model types including constraint‐based models, reaction‐diffusion models, logical network models, and rule‐based models. The format leverages two decades of SBML and a rich software ecosystem that transformed how systems biologists build and interact with models. More recently, the rise of multiscale models of whole cells and organs, and new data sources such as single‐cell measurements and live imaging, has precipitated new ways of integrating data with models. We provide our perspectives on the challenges presented by these developments and how SBML Level 3 provides the foundation needed to support this evolution.
- Published
- 2020
- Full Text
- View/download PDF
4. A kinase-deficient NTRK2 splice variant predominates in glioma and amplifies several oncogenic signaling pathways
- Author
-
Siobhan S. Pattwell, Sonali Arora, Patrick J. Cimino, Tatsuya Ozawa, Frank Szulzewsky, Pia Hoellerbauer, Tobias Bonifert, Benjamin G. Hoffstrom, Norman E. Boiani, Hamid Bolouri, Colin E. Correnti, Barbara Oldrini, John R. Silber, Massimo Squatrito, Patrick J. Paddison, and Eric C. Holland
- Subjects
Science - Abstract
Tropomyosin receptor kinase B (TrkB), encoded by the neurotrophic tyrosine receptor kinase 2 (NTRK2) gene, exhibits intricate splicing patterns and post-translational modifications. Here, the authors perform whole gene and transcript-level analyses and report the TrkB.T1 splice variant enhances PDGF-driven gliomas in vivo and augments PI3K signaling cascades in vitro.
- Published
- 2020
- Full Text
- View/download PDF
5. Multimodal analysis for human ex vivo studies shows extensive molecular changes from delays in blood processing
- Author
-
Adam K. Savage, Miriam V. Gutschow, Tony Chiang, Kathy Henderson, Richard Green, Monica Chaudhari, Elliott Swanson, Alexander T. Heubeck, Nina Kondza, Kelli C. Burley, Palak C. Genge, Cara Lord, Tanja Smith, Zachary Thomson, Aldan Beaubien, Ed Johnson, Jeff Goldy, Hamid Bolouri, Jane H. Buckner, Paul Meijer, Ernest M. Coffey, Peter J. Skene, Troy R. Torgerson, Xiao-jun Li, and Thomas F. Bumol
- Subjects
Molecular Physiology ,Immunology ,Proteomics ,Transcriptomics ,Science - Abstract
Summary: Multi-omic profiling of human peripheral blood is increasingly utilized to identify biomarkers and pathophysiologic mechanisms of disease. The importance of these platforms in clinical and translational studies led us to investigate the impact of delayed blood processing on the numbers and state of peripheral blood mononuclear cells (PBMC) and on the plasma proteome. Similar to previous studies, we show minimal effects of delayed processing on the numbers and general phenotype of PBMC up to 18 hours. In contrast, profound changes in the single-cell transcriptome and composition of the plasma proteome become evident as early as 6 hours after blood draw. These reflect patterns of cellular activation across diverse cell types that lead to progressive distancing of the gene expression state and plasma proteome from native in vivo biology. Differences accumulating during an overnight rest (18 hours) could confound relevant biologic variance related to many underlying disease states.
- Published
- 2021
- Full Text
- View/download PDF
6. A B-cell developmental gene regulatory network is activated in infant AML.
- Author
-
Hamid Bolouri, Rhonda Ries, Laura Pardo, Tiffany Hylkema, Wanding Zhou, Jenny L Smith, Amanda Leonti, Michael Loken, Jason E Farrar, Timothy J Triche, and Soheil Meshinchi
- Subjects
Medicine ,Science - Abstract
Infant Acute Myeloid Leukemia (AML) is a poorly-addressed, heterogeneous malignancy distinguished by surprisingly few mutations per patient but accompanied by myriad age-specific translocations. These characteristics make treatment of infant AML challenging. While infant AML is a relatively rare disease, it has enormous impact on families, and in terms of life-years-lost and life limiting morbidities. To better understand the mechanisms that drive infant AML, we performed integrative analyses of genome-wide mRNA, miRNA, and DNA-methylation data in diagnosis-stage patient samples. Here, we report the activation of an onco-fetal B-cell developmental gene regulatory network in infant AML. AML in infants is genomically distinct from AML in older children/adults in that it has more structural genomic aberrations and fewer mutations. Differential expression analysis of ~1500 pediatric AML samples revealed a large number of infant-specific genes, many of which are associated with B cell development and function. 18 of these genes form a well-studied B-cell gene regulatory network that includes the epigenetic regulators BRD4 and POU2AF1, and their onco-fetal targets LIN28B and IGF2BP3. All four genes are hypo-methylated in infant AML. Moreover, micro-RNA Let7a-2 is expressed in a mutually exclusive manner with its target and regulator LIN28B. These findings suggest infant AML may respond to bromodomain inhibitors and immune therapies targeting CD19, CD20, CD22, and CD79A.
- Published
- 2021
- Full Text
- View/download PDF
7. A De Novo Mouse Model of C11orf95-RELA Fusion-Driven Ependymoma Identifies Driver Functions in Addition to NF-κB
- Author
-
Tatsuya Ozawa, Sonali Arora, Frank Szulzewsky, Gordana Juric-Sekhar, Yoshiteru Miyajima, Hamid Bolouri, Yoshie Yasui, Jason Barber, Robert Kupp, James Dalton, Terreia S. Jones, Mitsutoshi Nakada, Toshihiro Kumabe, David W. Ellison, Richard J. Gilbertson, and Eric C. Holland
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: The majority of supratentorial ependymomas (ST-ependymomas) have few mutations but frequently display chromothripsis of chromosome 11q that generates a fusion between C11orf95 and RELA (RELAFUS). Neural stem cells transduced with RELAFUS ex vivo form ependymomas when implanted in the brain. These tumors display enhanced NF-κB signaling, suggesting that this aberrant signal is the principal mechanism of oncogenesis. However, it is not known whether RELAFUS is sufficient to drive de novo ependymoma tumorigenesis in the brain and, if so, whether these tumors also arise from neural stem cells. We show that RELAFUS drives ST-ependymoma formation from periventricular neural stem cells in mice and that RELAFUS-induced tumorigenesis is likely dependent on a series of cell signaling pathways in addition to NF-κB. : The C11orf95-RELA fusion (RELAFUS) has been found in a distinct subset of supratentorial ependymomas. Ozawa et al. show that RELAFUS is sufficient to drive ST-ependymoma formation from periventricular neural stem cells in mice. Furthermore, they show that RELAFUS-induced tumorigenesis might depend on other cell signaling pathways in addition to NF-κB. Keywords: ependymoma, fusion gene, NF-κB signaling, RCAS/tv-a system, mouse model, RELA, C11orf95
- Published
- 2018
- Full Text
- View/download PDF
8. Multidimensional scaling of diffuse gliomas: application to the 2016 World Health Organization classification system with prognostically relevant molecular subtype discovery
- Author
-
Patrick J. Cimino, Michael Zager, Lisa McFerrin, Hans-Georg Wirsching, Hamid Bolouri, Bettina Hentschel, Andreas von Deimling, David Jones, Guido Reifenberger, Michael Weller, and Eric C. Holland
- Subjects
Oncoscape ,Glioma ,Glioblastoma ,Astrocytoma ,Oligodendroglioma ,Isocitrate Dehydrogenase (IDH) ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Recent updating of the World Health Organization (WHO) classification of central nervous system (CNS) tumors in 2016 demonstrates the first organized effort to restructure brain tumor classification by incorporating histomorphologic features with recurrent molecular alterations. Revised CNS tumor diagnostic criteria also attempt to reduce interobserver variability of histological interpretation and provide more accurate stratification related to clinical outcome. As an example, diffuse gliomas (WHO grades II–IV) are now molecularly stratified based upon isocitrate dehydrogenase 1 or 2 (IDH) mutational status, with gliomas of WHO grades II and III being substratified according to 1p/19q codeletion status. For now, grading of diffuse gliomas is still dependent upon histological parameters. Independent of WHO classification criteria, multidimensional scaling analysis of molecular signatures for diffuse gliomas from The Cancer Genome Atlas (TCGA) has identified distinct molecular subgroups, and allows for their visualization in 2-dimensional (2D) space. Using the web-based platform Oncoscape as a tool, we applied multidimensional scaling-derived molecular groups to the 2D visualization of the 2016 WHO classification of diffuse gliomas. Here we show that molecular multidimensional scaling of TCGA data provides 2D clustering that represents the 2016 WHO classification of diffuse gliomas. Additionally, we used this platform to successfully identify and define novel copy-number alteration-based molecular subtypes, which are independent of WHO grading, as well as predictive of clinical outcome. The prognostic utility of these molecular subtypes was further validated using an independent data set of the German Glioma Network prospective glioblastoma patient cohort.
- Published
- 2017
- Full Text
- View/download PDF
9. Tables S1-S5 from Crebbp Loss Drives Small Cell Lung Cancer and Increases Sensitivity to HDAC Inhibition
- Author
-
David MacPherson, Kwon-Sik Park, Hamid Bolouri, Adi F. Gazdar, Smitha P.S. Pillai, Colin T. Dunn, Kee-Beom Kim, Ali H. Ibrahim, Nan Wu, Emily Eastwood, Dong-Wook Kim, Arnaud Augert, and Deshui Jia
- Abstract
Supplementary tables S1 to S5
- Published
- 2023
- Full Text
- View/download PDF
10. Figures S1-S16 from Crebbp Loss Drives Small Cell Lung Cancer and Increases Sensitivity to HDAC Inhibition
- Author
-
David MacPherson, Kwon-Sik Park, Hamid Bolouri, Adi F. Gazdar, Smitha P.S. Pillai, Colin T. Dunn, Kee-Beom Kim, Ali H. Ibrahim, Nan Wu, Emily Eastwood, Dong-Wook Kim, Arnaud Augert, and Deshui Jia
- Abstract
Supplementary Figures S1 to S16
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figures from Comprehensive Transcriptome Profiling of Cryptic CBFA2T3–GLIS2 Fusion–Positive AML Defines Novel Therapeutic Options: A COG and TARGET Pediatric AML Study
- Author
-
Soheil Meshinchi, Hamid Bolouri, Vivian G. Oehler, Michael R. Loken, Daoud Meerzaman, Cu Nguyen, Timothy J. Triche, Jason E. Farrar, E. Anders Kolb, Richard Aplenc, Alan S. Gamis, Amanda R. Leonti, Suzan Imren, Quy Le, Keith R. Loeb, Carrie L. Cummings, Laura Pardo, Lisa Eidenschink Brodersen, Marianne T. Santaguida, Robert B. Gerbing, Todd A. Alonzo, Tiffany Hylkema, Rhonda E. Ries, and Jenny L. Smith
- Abstract
Supplemental Figures including additional Kaplan-Meier plots, heatmaps of surface antigen expression and gene expression, and miRNA correlations.
- Published
- 2023
- Full Text
- View/download PDF
12. Data from Comprehensive Transcriptome Profiling of Cryptic CBFA2T3–GLIS2 Fusion–Positive AML Defines Novel Therapeutic Options: A COG and TARGET Pediatric AML Study
- Author
-
Soheil Meshinchi, Hamid Bolouri, Vivian G. Oehler, Michael R. Loken, Daoud Meerzaman, Cu Nguyen, Timothy J. Triche, Jason E. Farrar, E. Anders Kolb, Richard Aplenc, Alan S. Gamis, Amanda R. Leonti, Suzan Imren, Quy Le, Keith R. Loeb, Carrie L. Cummings, Laura Pardo, Lisa Eidenschink Brodersen, Marianne T. Santaguida, Robert B. Gerbing, Todd A. Alonzo, Tiffany Hylkema, Rhonda E. Ries, and Jenny L. Smith
- Abstract
Purpose:A cryptic inv(16)(p13.3q24.3) encoding the CBFA2T3–GLIS2 fusion is associated with poor outcome in infants with acute megakaryocytic leukemia. We aimed to broaden our understanding of the pathogenesis of this fusion through transcriptome profiling.Experimental Design:Available RNA from children and young adults with de novo acute myeloid leukemia (AML; N = 1,049) underwent transcriptome sequencing (mRNA and miRNA). Transcriptome profiles for those with the CBFA2T3–GLIS2 fusion (N = 24) and without (N = 1,025) were contrasted to define fusion-specific miRNAs, genes, and pathways. Clinical annotations defined distinct fusion-associated disease characteristics and outcomes.Results:The CBFA2T3–GLIS2 fusion was restricted to infants P < 0.001), and the presence of this fusion was highly associated with adverse outcome (P < 0.001) across all morphologic classifications. Further, there was a striking paucity of recurrent cooperating mutations, and transduction of cord blood stem cells with this fusion was sufficient for malignant transformation. CBFA2T3–GLIS2 positive cases displayed marked upregulation of genes with cell membrane/extracellular matrix localization potential, including NCAM1 and GABRE. Additionally, miRNA profiling revealed significant overexpression of mature miR-224 and miR-452, which are intronic miRNAs transcribed from the GABRE locus. Gene-set enrichment identified dysregulated Hippo, TGFβ, and hedgehog signaling, as well as NCAM1 (CD56) interaction pathways. Therapeutic targeting of fusion-positive leukemic cells with CD56-directed antibody–drug conjugate caused significant cytotoxicity in leukemic blasts.Conclusions:The CBFA2T3–GLIS2 fusion defines a highly refractory entity limited to infants that appears to be sufficient for malignant transformation. Transcriptome profiling elucidated several highly targetable genes and pathways, including the identification of CD56, providing a highly plausible target for therapeutic intervention.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Methods from Comprehensive Transcriptome Profiling of Cryptic CBFA2T3–GLIS2 Fusion–Positive AML Defines Novel Therapeutic Options: A COG and TARGET Pediatric AML Study
- Author
-
Soheil Meshinchi, Hamid Bolouri, Vivian G. Oehler, Michael R. Loken, Daoud Meerzaman, Cu Nguyen, Timothy J. Triche, Jason E. Farrar, E. Anders Kolb, Richard Aplenc, Alan S. Gamis, Amanda R. Leonti, Suzan Imren, Quy Le, Keith R. Loeb, Carrie L. Cummings, Laura Pardo, Lisa Eidenschink Brodersen, Marianne T. Santaguida, Robert B. Gerbing, Todd A. Alonzo, Tiffany Hylkema, Rhonda E. Ries, and Jenny L. Smith
- Abstract
Supplemental methods that provides additional detail on statistical methods and laboratory protocols and reagents.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Tables from Comprehensive Transcriptome Profiling of Cryptic CBFA2T3–GLIS2 Fusion–Positive AML Defines Novel Therapeutic Options: A COG and TARGET Pediatric AML Study
- Author
-
Soheil Meshinchi, Hamid Bolouri, Vivian G. Oehler, Michael R. Loken, Daoud Meerzaman, Cu Nguyen, Timothy J. Triche, Jason E. Farrar, E. Anders Kolb, Richard Aplenc, Alan S. Gamis, Amanda R. Leonti, Suzan Imren, Quy Le, Keith R. Loeb, Carrie L. Cummings, Laura Pardo, Lisa Eidenschink Brodersen, Marianne T. Santaguida, Robert B. Gerbing, Todd A. Alonzo, Tiffany Hylkema, Rhonda E. Ries, and Jenny L. Smith
- Abstract
Supplemental tables including clinical characteristics and differentially expressed gene lists.
- Published
- 2023
- Full Text
- View/download PDF
15. Object-oriented regression for building predictive models with high dimensional omics data from translational studies.
- Author
-
Lue Ping Zhao and Hamid Bolouri
- Published
- 2016
- Full Text
- View/download PDF
16. Genome-wide CRISPR-Cas9 Screens Reveal Loss of Redundancy between PKMYT1 and WEE1 in Glioblastoma Stem-like Cells
- Author
-
Chad M. Toledo, Yu Ding, Pia Hoellerbauer, Ryan J. Davis, Ryan Basom, Emily J. Girard, Eunjee Lee, Philip Corrin, Traver Hart, Hamid Bolouri, Jerry Davison, Qing Zhang, Justin Hardcastle, Bruce J. Aronow, Christopher L. Plaisier, Nitin S. Baliga, Jason Moffat, Qi Lin, Xiao-Nan Li, Do-Hyun Nam, Jeongwu Lee, Steven M. Pollard, Jun Zhu, Jeffery J. Delrow, Bruce E. Clurman, James M. Olson, and Patrick J. Paddison
- Subjects
CRISPR-Cas9 ,gene editing ,Glioblastoma ,PKMYT1 ,Myt1 ,WEE1 ,cancer therapeutics ,functional genomics ,Biology (General) ,QH301-705.5 - Abstract
To identify therapeutic targets for glioblastoma (GBM), we performed genome-wide CRISPR-Cas9 knockout (KO) screens in patient-derived GBM stem-like cells (GSCs) and human neural stem/progenitors (NSCs), non-neoplastic stem cell controls, for genes required for their in vitro growth. Surprisingly, the vast majority GSC-lethal hits were found outside of molecular networks commonly altered in GBM and GSCs (e.g., oncogenic drivers). In vitro and in vivo validation of GSC-specific targets revealed several strong hits, including the wee1-like kinase, PKMYT1/Myt1. Mechanistic studies demonstrated that PKMYT1 acts redundantly with WEE1 to inhibit cyclin B-CDK1 activity via CDK1-Y15 phosphorylation and to promote timely completion of mitosis in NSCs. However, in GSCs, this redundancy is lost, most likely as a result of oncogenic signaling, causing GBM-specific lethality.
- Published
- 2015
- Full Text
- View/download PDF
17. Oncogenic role of a developmentally regulatedNTRK2splice variant
- Author
-
Siobhan S. Pattwell, Sonali Arora, Nicholas Nuechterlein, Michael Zager, Keith R. Loeb, Patrick J. Cimino, Nikolas C. Holland, Noemi Reche-Ley, Hamid Bolouri, Damian A. Almiron Bonnin, Frank Szulzewsky, Vaishnavi V. Phadnis, Tatsuya Ozawa, Michael J. Wagner, Michael C. Haffner, Junyue Cao, Jay Shendure, and Eric C. Holland
- Subjects
Multidisciplinary - Abstract
Temporally regulated alternative splicing choices are vital for proper development, yet the wrong splice choice may be detrimental. Here, we highlight a previously unidentified role for the neurotrophin receptor splice variant TrkB.T1 in neurodevelopment, embryogenesis, transformation, and oncogenesis across multiple tumor types in humans and mice. TrkB.T1 is the predominantNTRK2isoform across embryonic organogenesis, and forced overexpression of this embryonic pattern causes multiple solid and nonsolid tumors in mice in the context of tumor suppressor loss. TrkB.T1 also emerges as the predominantNTRKisoform expressed in a wide range of adult and pediatric tumors, including those harboring tropomyosin receptor kinase fusions. Affinity purification–mass spectrometry proteomic analysis reveals distinct interactors with known developmental and oncogenic signaling pathways such as Wnt, transforming growth factor–β, Sonic Hedgehog, and Ras. From alterations in splicing factors to changes in gene expression, the discovery of isoform specific oncogenes with embryonic ancestry has the potential to shape the way we think about developmental systems and oncology.
- Published
- 2022
- Full Text
- View/download PDF
18. Towards Computational Neural Systems through Developmental Evolution.
- Author
-
Alistair G. Rust, Rod Adams, Stella J. George, and Hamid Bolouri
- Published
- 2001
- Full Text
- View/download PDF
19. Integration of 198 ChIP-seq Datasets Reveals Human cis-Regulatory Regions.
- Author
-
Hamid Bolouri and Walter L. Ruzzo
- Published
- 2012
- Full Text
- View/download PDF
20. Image Redundancy Reduction for Neural Network Classification Using Discrete Cosine Transforms.
- Author
-
Zhengjun Pan, Alistair G. Rust, and Hamid Bolouri
- Published
- 2000
- Full Text
- View/download PDF
21. Staged training of Neocognitron by evolutionary algorithms.
- Author
-
Zhengjun Pan, Theo Sabisch, Rod Adams, and Hamid Bolouri
- Published
- 1999
- Full Text
- View/download PDF
22. Variability in estimated gene expression among commonly used RNA-seq pipelines
- Author
-
Hamid Bolouri, Eric C. Holland, Sonali Arora, and Siobhan S. Pattwell
- Subjects
Differential expression analysis ,Receptor, ErbB-2 ,lcsh:Medicine ,RNA-Seq ,Computational biology ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Abundance (ecology) ,Neoplasms ,Exome Sequencing ,Gene expression ,Humans ,Transcriptomics ,lcsh:Science ,Gene ,Selection (genetic algorithm) ,030304 developmental biology ,Platelet-Derived Growth Factor ,Principal Component Analysis ,0303 health sciences ,Messenger RNA ,Multidisciplinary ,Sequence Analysis, RNA ,Gene Expression Profiling ,lcsh:R ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,Nuclear Proteins ,Splicing Factor U2AF ,Regression ,Neoplasm Proteins ,Data processing ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,CCAAT-Enhancer-Binding Proteins ,lcsh:Q ,Nucleophosmin - Abstract
RNA-sequencing data is widely used to identify disease biomarkers and therapeutic targets using numerical methods such as clustering, classification, regression, and differential expression analysis. Such approaches rely on the assumption that mRNA abundance estimates from RNA-seq are reliable estimates of true expression levels. Here, using data from five RNA-seq processing pipelines applied to 6,690 human tumor and normal tissues, we show that nearly 88% of protein-coding genes have similar gene expression profiles across all pipelines. However, for >12% of protein-coding genes, current best-in-class RNA-seq processing pipelines differ in their abundance estimates by more than four-fold when applied to exactly the same samples and the same set of RNA-seq reads. Expression fold changes are similarly affected. Many of the impacted genes are widely studied disease-associated genes. We show that impacted genes exhibit diverse patterns of discordance among pipelines, suggesting that many inter-pipeline differences contribute to overall uncertainty in mRNA abundance estimates. A concerted, community-wide effort will be needed to develop gold-standards for estimating the mRNA abundance of the discordant genes reported here. In the meantime, our list of discordantly evaluated genes provides an important resource for robust marker discovery and target selection.
- Published
- 2020
- Full Text
- View/download PDF
23. Comprehensive Transcriptome Profiling of Cryptic CBFA2T3–GLIS2 Fusion–Positive AML Defines Novel Therapeutic Options: A COG and TARGET Pediatric AML Study
- Author
-
Keith R. Loeb, Richard Aplenc, Robert B. Gerbing, E. Anders Kolb, Amanda R. Leonti, Hamid Bolouri, Laura Pardo, Alan S. Gamis, Rhonda E. Ries, Todd A. Alonzo, Jenny L. Smith, Soheil Meshinchi, Suzan Imren, Lisa Eidenschink Brodersen, Vivian G. Oehler, Timothy J. Triche, Cu Nguyen, Jason E. Farrar, Carrie L. Cummings, Michael R. Loken, Daoud Meerzaman, Tiffany A. Hylkema, Quy Le, and Marianne Santaguida
- Subjects
0301 basic medicine ,Cancer Research ,RNA ,Myeloid leukemia ,Computational biology ,Biology ,medicine.disease ,Malignant transformation ,Pathogenesis ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,0302 clinical medicine ,Cog ,Oncology ,030220 oncology & carcinogenesis ,microRNA ,medicine ,Gene - Abstract
Purpose: A cryptic inv(16)(p13.3q24.3) encoding the CBFA2T3–GLIS2 fusion is associated with poor outcome in infants with acute megakaryocytic leukemia. We aimed to broaden our understanding of the pathogenesis of this fusion through transcriptome profiling. Experimental Design: Available RNA from children and young adults with de novo acute myeloid leukemia (AML; N = 1,049) underwent transcriptome sequencing (mRNA and miRNA). Transcriptome profiles for those with the CBFA2T3–GLIS2 fusion (N = 24) and without (N = 1,025) were contrasted to define fusion-specific miRNAs, genes, and pathways. Clinical annotations defined distinct fusion-associated disease characteristics and outcomes. Results: The CBFA2T3–GLIS2 fusion was restricted to infants Conclusions: The CBFA2T3–GLIS2 fusion defines a highly refractory entity limited to infants that appears to be sufficient for malignant transformation. Transcriptome profiling elucidated several highly targetable genes and pathways, including the identification of CD56, providing a highly plausible target for therapeutic intervention.
- Published
- 2020
- Full Text
- View/download PDF
24. Molecular Self-Organization in the Development Model for the Evolution of Large-scale Artificial Neural Networks.
- Author
-
Hamid Bolouri, Rod Adams, Stella J. George, and Alistair G. Rust
- Published
- 1998
25. Rotation, Translation, and Scaling Tolerant Recognition of Complex Shapes Using a Hierarchical Self-Organising Neural Network.
- Author
-
Theo Sabisch, Alistair Ferguson, and Hamid Bolouri
- Published
- 1997
26. Designing Development Rules for Artificial Evolution.
- Author
-
Alistair G. Rust, Rod Adams, Stella J. George, and Hamid Bolouri
- Published
- 1997
- Full Text
- View/download PDF
27. High-Speed Airborne Particle Monitoring Using Artificial Neural Networks.
- Author
-
Alistair Ferguson, Theo Sabisch, Paul Kaye, Laurence C. Dixon, and Hamid Bolouri
- Published
- 1995
28. A method for estimating stochastic noise in large genetic regulatory networks.
- Author
-
David Orrell, Stephen Ramsey, Pedro de Atauri, and Hamid Bolouri
- Published
- 2005
- Full Text
- View/download PDF
29. Dizzy: Stochastic Simulation of Large-scale Genetic Regulatory Networks.
- Author
-
Stephen Ramsey, David Orrell, and Hamid Bolouri
- Published
- 2005
- Full Text
- View/download PDF
30. A finite state automaton model for multi-neuron simulations.
- Author
-
Maria J. Schilstra, Alistair G. Rust, Rod Adams, and Hamid Bolouri
- Published
- 2002
- Full Text
- View/download PDF
31. Computational Modeling of Gene Regulatory Networks — A Primer
- Author
-
Hamid Bolouri
- Published
- 2008
32. Identification of complex shapes using a self organizing neural system.
- Author
-
Theo Sabisch, Alistair Ferguson, and Hamid Bolouri
- Published
- 2000
- Full Text
- View/download PDF
33. Improving Reinforcement Learning in Stochastic {RAM}-Based Neural Networks.
- Author
-
Alistair Ferguson and Hamid Bolouri
- Published
- 1996
- Full Text
- View/download PDF
34. Multimodal analysis for human ex vivo studies shows extensive molecular changes from delays in blood processing
- Author
-
Kelli C. Burley, Hamid Bolouri, Jeff Goldy, Paul Meijer, Peter J Skene, Ed Johnson, Thomas F. Bumol, Tanja Smith, Elliott Swanson, Cara Lord, Miriam V. Gutschow, Aldan Beaubien, Alexander T. Heubeck, Zachary Thomson, Ernest M. Coffey, Adam K. Savage, Xiao-jun Li, Monica Chaudhari, Jane H. Buckner, Tony Chiang, Palak C Genge, Nina Kondza, Kathy Henderson, Richard Green, and Troy R. Torgerson
- Subjects
0301 basic medicine ,Proteomics ,Cell type ,Multidisciplinary ,Science ,Immunology ,02 engineering and technology ,Biology ,021001 nanoscience & nanotechnology ,Peripheral blood mononuclear cell ,Phenotype ,Article ,Molecular Physiology ,Transcriptome ,03 medical and health sciences ,030104 developmental biology ,In vivo ,Gene expression ,Proteome ,0210 nano-technology ,Transcriptomics ,Ex vivo - Abstract
Summary Multi-omic profiling of human peripheral blood is increasingly utilized to identify biomarkers and pathophysiologic mechanisms of disease. The importance of these platforms in clinical and translational studies led us to investigate the impact of delayed blood processing on the numbers and state of peripheral blood mononuclear cells (PBMC) and on the plasma proteome. Similar to previous studies, we show minimal effects of delayed processing on the numbers and general phenotype of PBMC up to 18 hours. In contrast, profound changes in the single-cell transcriptome and composition of the plasma proteome become evident as early as 6 hours after blood draw. These reflect patterns of cellular activation across diverse cell types that lead to progressive distancing of the gene expression state and plasma proteome from native in vivo biology. Differences accumulating during an overnight rest (18 hours) could confound relevant biologic variance related to many underlying disease states., Graphical abstract, Highlights • Studies of human blood cells and plasma are highly sensitive to process variability • Time variability distorts biology in cutting-edge single-cell and multiplex assays • Longitudinal, multi-modal, and aligned data enable data qualification and exploration • Dataset holds potential novel, multi-modal biological correlations and hypotheses, Molecular Physiology ; Immunology ; Proteomics ; Transcriptomics
- Published
- 2021
35. The COVID-19 immune landscape is dynamically and reversibly correlated with disease severity
- Author
-
Cate Speake, Daniel J. Campbell, Hamid Bolouri, Anne M. Hocking, Jane H. Buckner, David A. G. Skibinski, Uma Malhotra, S. Alice Long, and Jessica A. Hamerman
- Subjects
Adult ,Male ,0301 basic medicine ,Coronavirus disease 2019 (COVID-19) ,Disease ,Adaptive Immunity ,Severity of Illness Index ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Disease severity ,Humans ,Medicine ,Mass cytometry ,Whole blood ,Aged ,Aged, 80 and over ,SARS-CoV-2 ,business.industry ,Critically ill ,COVID-19 ,General Medicine ,Middle Aged ,biochemical phenomena, metabolism, and nutrition ,Flow Cytometry ,Immunity, Innate ,Blockade ,COVID-19 Drug Treatment ,030104 developmental biology ,Disease Presentation ,030220 oncology & carcinogenesis ,Cohort ,Immunology ,Female ,Clinical Medicine ,business - Abstract
BACKGROUND: Despite a rapidly growing body of literature on coronavirus disease 2019 (COVID-19), our understanding of the immune correlates of disease severity, course, and outcome remains poor. METHODS: Using mass cytometry, we assessed the immune landscape in longitudinal whole-blood specimens from 59 patients presenting with acute COVID-19 and classified based on maximal disease severity. Hospitalized patients negative for SARS-CoV-2 were used as controls. RESULTS: We found that the immune landscape in COVID-19 formed 3 dominant clusters, which correlated with disease severity. Longitudinal analysis identified a pattern of productive innate and adaptive immune responses in individuals who had a moderate disease course, whereas those with severe disease had features suggestive of a protracted and dysregulated immune response. Further, we identified coordinate immune alterations accompanying clinical improvement and decline that were also seen in patients who received IL-6 pathway blockade. CONCLUSION: The hospitalized COVID-19 negative cohort allowed us to identify immune alterations that were shared between severe COVID-19 and other critically ill patients. Collectively, our findings indicate that selection of immune interventions should be based in part on disease presentation and early disease trajectory due to the profound differences in the immune response in those with mild to moderate disease and those with the most severe disease. FUNDING: Benaroya Family Foundation, the Leonard and Norma Klorfine Foundation, Glenn and Mary Lynn Mounger, and the National Institutes of Health.
- Published
- 2021
- Full Text
- View/download PDF
36. A B-cell developmental gene regulatory network is activated in infant AML
- Author
-
Wanding Zhou, Timothy J. Triche, Rhonda E. Ries, Jenny L. Smith, Amanda R. Leonti, Soheil Meshinchi, Laura Pardo, Michael R. Loken, Jason E. Farrar, Hamid Bolouri, and Tiffany A. Hylkema
- Subjects
B Cells ,Gene regulatory network ,Cell Cycle Proteins ,Biochemistry ,Pediatrics ,Hematologic Cancers and Related Disorders ,Families ,White Blood Cells ,Animal Cells ,hemic and lymphatic diseases ,Medicine and Health Sciences ,Gene Regulatory Networks ,Children ,B-Lymphocytes ,Multidisciplinary ,Leukemia ,DNA methylation ,biology ,Myeloid leukemia ,RNA-Binding Proteins ,Hematology ,Genomics ,Myeloid Leukemia ,Chromatin ,Up-Regulation ,Nucleic acids ,Leukemia, Myeloid, Acute ,Oncology ,Medicine ,Epigenetics ,Cellular Types ,DNA modification ,Infants ,Chromatin modification ,Research Article ,Chromosome biology ,Acute Myeloid Leukemia ,BRD4 ,Immune Cells ,Science ,Immunology ,CD19 ,microRNA ,Genetics ,Humans ,RNA, Messenger ,Antibody-Producing Cells ,Non-coding RNA ,Gene ,Natural antisense transcripts ,Blood Cells ,Infant ,Cancers and Neoplasms ,Biology and Life Sciences ,Cell Biology ,DNA ,Gene regulation ,MicroRNAs ,Age Groups ,People and Places ,biology.protein ,Cancer research ,Trans-Activators ,RNA ,Population Groupings ,Gene expression ,Transcription Factors - Abstract
Infant Acute Myeloid Leukemia (AML) is a poorly-addressed, heterogeneous malignancy distinguished by surprisingly few mutations per patient but accompanied by myriad age-specific translocations. These characteristics make treatment of infant AML challenging. While infant AML is a relatively rare disease, it has enormous impact on families, and in terms of life-years-lost and life limiting morbidities. To better understand the mechanisms that drive infant AML, we performed integrative analyses of genome-wide mRNA, miRNA, and DNA-methylation data in diagnosis-stage patient samples. Here, we report the activation of an onco-fetal B-cell developmental gene regulatory network in infant AML. AML in infants is genomically distinct from AML in older children/adults in that it has more structural genomic aberrations and fewer mutations. Differential expression analysis of ~1500 pediatric AML samples revealed a large number of infant-specific genes, many of which are associated with B cell development and function. 18 of these genes form a well-studied B-cell gene regulatory network that includes the epigenetic regulators BRD4 and POU2AF1, and their onco-fetal targets LIN28B and IGF2BP3. All four genes are hypo-methylated in infant AML. Moreover, micro-RNA Let7a-2 is expressed in a mutually exclusive manner with its target and regulator LIN28B. These findings suggest infant AML may respond to bromodomain inhibitors and immune therapies targeting CD19, CD20, CD22, and CD79A.
- Published
- 2021
37. A kinase-deficient NTRK2 splice variant predominates in glioma and amplifies several oncogenic signaling pathways
- Author
-
Patrick J. Cimino, Sonali Arora, Tobias Bonifert, Norman Boiani, Tatsuya Ozawa, Barbara Oldrini, Frank Szulzewsky, Eric C. Holland, Hamid Bolouri, Massimo Squatrito, Siobhan S. Pattwell, Colin Correnti, Patrick J. Paddison, Pia Hoellerbauer, Benjamin G. Hoffstrom, John R. Silber, United States Department of Health & Human Services National Institutes of Health (NIH) - USA, American Cancer Society, French National Research Agency (ANR), Alzheimer's Disease Research Center, United States of Department of Health & Human Services, and French Development Agency
- Subjects
0301 basic medicine ,RNA splicing ,Oncogene Proteins, Fusion ,Carcinogenesis ,RNA Splicing ,Science ,General Physics and Astronomy ,Tropomyosin receptor kinase B ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Fusion gene ,03 medical and health sciences ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Neural Stem Cells ,RNA Isoforms ,Animals ,Humans ,Receptor, trkB ,splice ,lcsh:Science ,Protein kinase B ,Cells, Cultured ,Multidisciplinary ,Membrane Glycoproteins ,Brain Neoplasms ,Gene Expression Profiling ,Alternative splicing ,Brain ,High-Throughput Nucleotide Sequencing ,Neurotrophic factors ,General Chemistry ,Glioma ,Oncogenes ,Cell biology ,Gene expression profiling ,CNS cancer ,030104 developmental biology ,Gene Ontology ,nervous system ,030220 oncology & carcinogenesis ,NIH 3T3 Cells ,lcsh:Q ,Signal transduction ,Signal Transduction - Abstract
Independent scientific achievements have led to the discovery of aberrant splicing patterns in oncogenesis, while more recent advances have uncovered novel gene fusions involving neurotrophic tyrosine receptor kinases (NTRKs) in gliomas. The exploration of NTRK splice variants in normal and neoplastic brain provides an intersection of these two rapidly evolving fields. Tropomyosin receptor kinase B (TrkB), encoded NTRK2, is known for critical roles in neuronal survival, differentiation, molecular properties associated with memory, and exhibits intricate splicing patterns and post-translational modifications. Here, we show a role for a truncated NTRK2 splice variant, TrkB.T1, in human glioma. TrkB.T1 enhances PDGF-driven gliomas in vivo, augments PDGF-induced Akt and STAT3 signaling in vitro, while next generation sequencing broadly implicates TrkB.T1 in the PI3K signaling cascades in a ligand-independent fashion. These TrkB.T1 findings highlight the importance of expanding upon whole gene and gene fusion analyses to include splice variants in basic and translational neuro-oncology research., Tropomyosin receptor kinase B (TrkB), encoded by the neurotrophic tyrosine receptor kinase 2 (NTRK2) gene, exhibits intricate splicing patterns and post-translational modifications. Here, the authors perform whole gene and transcript-level analyses and report the TrkB.T1 splice variant enhances PDGF-driven gliomas in vivo and augments PI3K signaling cascades in vitro.
- Published
- 2020
38. Copy number profiling across glioblastoma populations has implications for clinical trial design
- Author
-
Lisa McFerrin, Eric C. Holland, Patrick J. Cimino, Sonali Arora, Hans-Georg Wirsching, Raul Rabadan, Michael Weller, and Hamid Bolouri
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Clinical study design ,Population ,medicine.disease ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,Glioma ,Internal medicine ,Cohort ,Medicine ,Neurology (clinical) ,business ,education ,Survival rate ,Exome ,Cohort study - Abstract
Background Copy number alterations form prognostic molecular subtypes of glioblastoma with clear differences in median overall survival. In this study, we leverage molecular data from several glioblastoma cohorts to define the distribution of copy number subtypes across random cohorts as well as cohorts with selection biases for patients with inherently better outcome. Methods Copy number subtype frequency was established for 4 glioblastoma patient cohorts. Two randomly selected cohorts include The Cancer Genome Atlas (TCGA) and the German Glioma Network (GGN). Two more selective cohorts include the phase II trial ARTE in elderly patients with newly diagnosed glioblastoma and a multi-institutional cohort focused on paired resected initial/recurrent glioblastoma. The paired initial/recurrent cohort also had exome data available, which allowed for evaluation of multidimensional scaling analysis. Results Smaller selective glioblastoma cohorts are enriched for copy number subtypes that are associated with better survival, reflecting the selection of patients who do well enough to enter a clinical trial or who are deemed well enough to undergo resection at recurrence. Adding exome data to copy number data provides additional data reflective of outcome. Conclusions The overall outcome for diffuse glioma patients is predicted by DNA structure at initial tumor resection. Molecular signature shifts across glioblastoma populations reflect the inherent bias of patient selection toward longer survival in clinical trials. Therefore it may be important to include molecular profiling, including copy number, when enrolling patients for clinical trials in order to balance arms and extrapolate relevance to the general glioblastoma population.
- Published
- 2018
- Full Text
- View/download PDF
39. A De Novo Mouse Model of C11orf95-RELA Fusion-Driven Ependymoma Identifies Driver Functions in Addition to NF-κB
- Author
-
Mitsutoshi Nakada, Toshihiro Kumabe, Jason Barber, Frank Szulzewsky, Yoshie Yasui, Gordana Juric-Sekhar, James Dalton, Hamid Bolouri, Robert Kupp, Richard J. Gilbertson, Yoshiteru Miyajima, Eric C. Holland, Tatsuya Ozawa, Terreia S. Jones, David W. Ellison, Sonali Arora, Gilbertson, Richard [0000-0001-7539-9472], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Ependymoma ,mouse model ,RELA ,Biology ,RCAS/tv-a system ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,Fusion gene ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Neural Stem Cells ,medicine ,Animals ,Humans ,Oncogene Fusion ,lcsh:QH301-705.5 ,Principal Component Analysis ,Chromothripsis ,Brain Neoplasms ,C11orf95 ,NF-kappa B ,Transcription Factor RelA ,Chromosome ,Proteins ,Supratentorial Neoplasms ,NF-κB ,medicine.disease ,Neural stem cell ,DNA-Binding Proteins ,Disease Models, Animal ,030104 developmental biology ,Cell Transformation, Neoplastic ,chemistry ,fusion gene ,lcsh:Biology (General) ,NF-κB signaling ,Cancer research ,Carcinogenesis ,Transcriptome ,Cell signaling pathways ,Signal Transduction - Abstract
SUMMARY The majority of supratentorial ependymomas (ST-ependymomas) have few mutations but frequently display chromothripsis of chromosome 11q that generates a fusion between C11orf95 and RELA (RELAFUS). Neural stem cells transduced with RELAFUS ex vivo form ependymomas when implanted in the brain. These tumors display enhanced NF-κB signaling, suggesting that this aberrant signal is the principal mechanism of oncogenesis. However, it is not known whether RELAFUS is sufficient to drive de novo ependymoma tumorigenesis in the brain and, if so, whether these tumors also arise from neural stem cells. We show that RELAFUS drives ST-ependymoma formation from periventricular neural stem cells in mice and that RELAFUS-induced tumorigenesis is likely dependent on a series of cell signaling pathways in addition to NF-κB., Graphical Abstract, In Brief The C11orf95-RELA fusion (RELAFUS) has been found in a distinct subset of supratentorial ependymomas. Ozawa et al. show that RELAFUS is sufficient to drive ST-ependymoma formation from periventricular neural stem cells in mice. Furthermore, they show that RELAFUS-induced tumorigenesis might depend on other cell signaling pathways in addition to NF-κB.
- Published
- 2018
40. TMOD-30. NTRK2 SPLICE VARIANT, TRKB.T1, LINKS NEUROBIOLOGY, EMBRYONIC DEVELOPMENT, AND ONCOGENESIS
- Author
-
Patrick J. Cimino, Tatsuya Ozawa, Siobhan S. Pattwell, Eric C. Holland, Sonali Arora, Nicholas Nuechterlein, Michael Zager, Junyue Cao, Nikolas Holland, Hamid Bolouri, Keith R. Loeb, Jay Shendure, and Michael C. Haffner
- Subjects
Cancer Research ,nervous system ,Oncology ,Embryogenesis ,Alternative splicing ,medicine ,Neurology (clinical) ,Biology ,Carcinogenesis ,medicine.disease_cause ,Neuroscience - Abstract
Temporally regulated alternative splicing choices are vital for proper development yet the wrong splice choice may be detrimental. Here we highlight a novel role for the neurotrophin receptor splice variant TrkB.T1 in neurodevelopment, embryogenesis, transformation, and oncogenesis across multiple tumor types in both humans and mice. TrkB.T1 is the predominant NTRK2 isoform across embryonic organogenesis and is highly expressed in a wide range of adult and pediatric tumors. Further, forced expression of TrkB.T1 causes multiple solid and non-solid tumors in mice in the context of tumor suppressor loss. These results highlight a unique role for the neurotrophin receptor splicing in development and oncogenesis and underscore the need for considering alternative splicing and transcript level data in neuroscience, developmental biology, and oncology research.
- Published
- 2021
- Full Text
- View/download PDF
41. Menu-driven cloud computing and resource sharing for R and Bioconductor.
- Author
-
Hamid Bolouri, Rajiv Dulepet, and Michael Angerman
- Published
- 2011
- Full Text
- View/download PDF
42. SBML Level 3: an extensible format for the exchange and reuse of biological models
- Author
-
Edda Klipp, Marco Antoniotti, Frank Bergmann, James C. Schaff, Peter D. Karp, Daniel Lucio, Kedar Nath Natarajan, Thomas M. Hamm, Leandro Watanabe, Henning Hermjakob, David Tolnay, John Wagner, Joerg Stelling, Alida Palmisano, Falk Schreiber, Yukiko Matsuoka, Harold F. Gómez, Huaiyu Mi, Carole J. Proctor, Ulrike Wittig, Neil Swainston, Jan Červený, Denis Thieffry, Piero Dalle Pezze, Julio Saez-Rodriguez, Maciej J. Swat, Bin Hu, Martina Kutmon, Thomas Pfau, Bas Teusink, Sarah M. Keating, Fedor A. Kolpakov, Andreas Dräger, Pedro Mendes, Martin Scharm, Emek Demir, Ioannis Xenarios, Christoph Flamm, Axel von Kamp, Darren J. Wilkinson, Nick Juty, Fengkai Zhang, Leonard A. Harris, Michael Schubert, Dagmar Waltemath, Lucian P. Smith, Steffen Klamt, Herbert M. Sauro, Ali Ebrahim, Wolfram Liebermeister, Christian Knüpfer, Nicolas Rodriguez, Tramy Nguyen, Naoki Tanimura, Christopher Cox, Stuart C. Sealfon, Nicholas Alexander Allen, Clemens Wrzodek, Bastian R. Angermann, Martin Meier-Schellersheim, Anna Zhukova, Jean-Baptiste Pettit, Hovakim Grabski, Devin P. Sullivan, Claudine Chaouiya, Michael L. Blinov, John Doyle, Ilya Kiselev, Roman Schulte, Alex Gutteridge, Mélanie Courtot, Eric Mjolsness, Finja Wrzodek, Rahuman S Malik-Sheriff, Ronan M. T. Fleming, Bruce E. Shapiro, Kimberly Begley, Leslie M. Loew, Colin S. Gillespie, Ibrahim Vazirabad, Michael Hucka, Akira Funahashi, Bernhard O. Palsson, Hamid Bolouri, Tomáš Helikar, Camille Laibe, William S. Denney, Chris T. Evelo, Florian Mittag, William S. Hlavacek, Ron Henkel, Harish Dharuri, Julien Dorier, Karthik Raman, Martina Fröhlich, Conor Lawless, Rainer Machné, Falko Krause, Damon Hachmeister, Matthias König, Clifford A. Shaffer, Benjamin D. Heavner, Douglas B. Kell, Jonathan R. Karr, Mihai Glont, Lukas Endler, Melanie I. Stefan, Robert Phair, Lu Li, Henning Schmidt, Dirk Drasdo, Johan Elf, Allyson L. Lister, Hiroaki Kitano, Richard R. Adams, Oliver A. Ruebenacker, Roland Keller, Sven Sahle, Ion I. Moraru, Gary D. Bader, Poul M. F. Nielsen, Johann M. Rohwer, Johannes Eichner, Daniel R. Hyduke, James R. Faeder, Stefan Hoops, Emanuel Gonçalves, Yuichiro Inagaki, Aurélien Naldi, Koichi Takahashi, Sylvain Soliman, Brett G. Olivier, Kieran Smallbone, Stuart L. Moodie, Pedro T. Monteiro, Chris J. Myers, Martin Golebiewski, Tomas Radivoyevitch, Jeremy Zucker, Hidde de Jong, Andrew Finney, Keating, S, Waltemath, D, König, M, Zhang, F, Dräger, A, Chaouiya, C, Bergmann, F, Finney, A, Gillespie, C, Helikar, T, Hoops, S, Malik-Sheriff, R, Moodie, S, Moraru, I, Myers, C, Naldi, A, Olivier, B, Sahle, S, Schaff, J, Smith, L, Swat, M, Thieffry, D, Watanabe, L, Wilkinson, D, Blinov, M, Begley, K, Faeder, J, Gómez, H, Hamm, T, Inagaki, Y, Liebermeister, W, Lister, A, Lucio, D, Mjolsness, E, Proctor, C, Raman, K, Rodriguez, N, Shaffer, C, Shapiro, B, Stelling, J, Swainston, N, Tanimura, N, Wagner, J, Meier-Schellersheim, M, Sauro, H, Palsson, B, Bolouri, H, Kitano, H, Funahashi, A, Hermjakob, H, Doyle, J, Hucka, M, Adams, R, Allen, N, Angermann, B, Antoniotti, M, Bader, G, Červený, J, Courtot, M, Cox, C, Dalle Pezze, P, Demir, E, Denney, W, Dharuri, H, Dorier, J, Drasdo, D, Ebrahim, A, Eichner, J, Elf, J, Endler, L, Evelo, C, Flamm, C, Fleming, R, Fröhlich, M, Glont, M, Gonçalves, E, Golebiewski, M, Grabski, H, Gutteridge, A, Hachmeister, D, Harris, L, Heavner, B, Henkel, R, Hlavacek, W, Hu, B, Hyduke, D, Jong, H, Juty, N, Karp, P, Karr, J, Kell, D, Keller, R, Kiselev, I, Klamt, S, Klipp, E, Knüpfer, C, Kolpakov, F, Krause, F, Kutmon, M, Laibe, C, Lawless, C, Li, L, Loew, L, Machne, R, Matsuoka, Y, Mendes, P, Mi, H, Mittag, F, Monteiro, P, Natarajan, K, Nielsen, P, Nguyen, T, Palmisano, A, Jean-Baptiste, P, Pfau, T, Phair, R, Radivoyevitch, T, Rohwer, J, Ruebenacker, O, Saez-Rodriguez, J, Scharm, M, Schmidt, H, Schreiber, F, Schubert, M, Schulte, R, Sealfon, S, Smallbone, K, Soliman, S, Stefan, M, Sullivan, D, Takahashi, K, Teusink, B, Tolnay, D, Vazirabad, I, Kamp, A, Wittig, U, Wrzodek, C, Wrzodek, F, Xenarios, I, Zhukova, A, Zucker, J, European Bioinformatics Institute [Hinxton] (EMBL-EBI), EMBL Heidelberg, Heidelberg University Hospital [Heidelberg], Swiss Institute of Bioinformatics [Lausanne] (SIB), Université de Lausanne = University of Lausanne (UNIL), European Molecular Biology Laboratory (EMBL), University of Connecticut (UCONN), National Institutes of Health [Bethesda] (NIH), Chercheur indépendant, Amazon Web Services [Seattle] (AWS), Università degli Studi di Milano-Bicocca = University of Milano-Bicocca (UNIMIB), University of Toronto, Masaryk University [Brno] (MUNI), Terry Fox Laboratory, BC Cancer Agency (BCCRC)-British Columbia Cancer Agency Research Centre, The University of Tennessee [Knoxville], The Babraham Institute [Cambridge, UK], Oregon Health and Science University [Portland] (OHSU), Human Predictions LLC, Illumina, Swiss-Prot Group, Swiss Institute of Bioinformatics [Genève] (SIB), Modelling and Analysis for Medical and Biological Applications (MAMBA), Inria de Paris, Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria)-Laboratoire Jacques-Louis Lions (LJLL (UMR_7598)), Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), University of California [San Diego] (UC San Diego), University of California (UC), Center for Bioinformatics (ZBIT), Eberhard Karls Universität Tübingen = Eberhard Karls University of Tuebingen, Uppsala University, Institut für Populationsgenetik [Vienna], Veterinärmedizinische Universität Wien, Maastricht University [Maastricht], Alpen-Adria-Universität Klagenfurt [Klagenfurt, Austria], Medizinische Universität Wien = Medical University of Vienna, German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ), Heidelberg Institute for Theoretical Studies (HITS ), Russian-Armenian University (RAU), GlaxoSmithKline [Stevenage, UK] (GSK), GlaxoSmithKline [Headquarters, London, UK] (GSK), Microsoft Technology Licensing (MTL), Microsoft Corporation [Redmond, Wash.], Vanderbilt University School of Medicine [Nashville], University of Washington [Seattle], University of Rostock, Los Alamos National Laboratory (LANL), Lorentz Institute, Universiteit Leiden, Tegmine Therapeutics, Modeling, simulation, measurement, and control of bacterial regulatory networks (IBIS), Laboratoire Adaptation et pathogénie des micro-organismes [Grenoble] (LAPM), Université Joseph Fourier - Grenoble 1 (UJF)-Centre National de la Recherche Scientifique (CNRS)-Université Joseph Fourier - Grenoble 1 (UJF)-Centre National de la Recherche Scientifique (CNRS)-Inria Grenoble - Rhône-Alpes, Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria)-Institut Jean Roget, SRI International [Menlo Park] (SRI), Icahn School of Medicine at Mount Sinai [New York] (MSSM), University of Liverpool, Universitätsklinikum Tübingen - University Hospital of Tübingen, Institute of Information and Computational Technologies (IICT), Max Planck Institute for Dynamics of Complex Technical Systems, Max-Planck-Gesellschaft, Max-Planck-Institut für Molekulare Genetik (MPIMG), Friedrich-Schiller-Universität = Friedrich Schiller University Jena [Jena, Germany], Humboldt University Of Berlin, Newcastle University [Newcastle], École polytechnique (X), Heinrich Heine Universität Düsseldorf = Heinrich Heine University [Düsseldorf], The Systems Biology Institute [Tokyo] (SBI), Centro de Quimica Estrutural (CQE), Instituto Superior Técnico, Universidade Técnica de Lisboa (IST), University of Southern California (USC), Instituto Gulbenkian de Ciência [Oeiras] (IGC), Fundação Calouste Gulbenkian, University of Southern Denmark (SDU), University of Auckland [Auckland], University of Utah, Virginia Tech [Blacksburg], University of Luxembourg [Luxembourg], Integrative Bioinformatics Inc [Mountain View], Cleveland Clinic, Stellenbosch University, Broad Institute of MIT and Harvard (BROAD INSTITUTE), Harvard Medical School [Boston] (HMS)-Massachusetts Institute of Technology (MIT)-Massachusetts General Hospital [Boston], Universität Heidelberg [Heidelberg] = Heidelberg University, Leibniz Institute of Plant Genetics and Crop Plant Research [Gatersleben] (IPK-Gatersleben), Laboratoire de Biologie du Développement de Villefranche sur mer (LBDV), Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut de la Mer de Villefranche (IMEV), Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Mount Sinai School of Medicine, Department of Psychiatry-Icahn School of Medicine at Mount Sinai [New York] (MSSM), University of Manchester [Manchester], Computational systems biology and optimization (Lifeware), Inria Saclay - Ile de France, Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria), California Institute of Technology (CALTECH), Encodia Inc [San Diego], Shinshu University [Nagano], University of Amsterdam [Amsterdam] (UvA), Versiti Blood Center of Wisconsin, Greifswald University Hospital, Bioinformatique évolutive - Evolutionary Bioinformatics, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Pacific Northwest National Laboratory (PNNL), National Institute of Allergy and Infectious Diseases [Bethesda] (NIAID-NIH), Department of Bioengineering, University of California (UC)-University of California (UC), ANSYS, Virginia Polytechnic Institute and State University [Blacksburg], Eight Pillars Ltd, Center for Integrative Genomics - Institute of Bioinformatics, Génopode (CIG), Université de Lausanne = University of Lausanne (UNIL)-Université de Lausanne = University of Lausanne (UNIL), Universität Heidelberg, Bioquant, Applied Biomathematics [New York], SimCYP Ltd, Institut de biologie de l'ENS Paris (IBENS), Département de Biologie - ENS Paris, École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), University of Utah School of Medicine [Salt Lake City], University of Pittsburgh School of Medicine, Pennsylvania Commonwealth System of Higher Education (PCSHE), Eidgenössische Technische Hochschule - Swiss Federal Institute of Technology [Zürich] (ETH Zürich), Mizuho Information and Research Institute, Mathématiques et Informatique Appliquées du Génome à l'Environnement [Jouy-En-Josas] (MaIAGE), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), University of Oxford, Computer Science (North Carolina State University), North Carolina State University [Raleigh] (NC State), University of North Carolina System (UNC)-University of North Carolina System (UNC), University of California [Irvine] (UC Irvine), Indian Institute of Technology Madras (IIT Madras), California State University [Northridge] (CSUN), Biotechnology and Biological Sciences Research Council (BBSRC), IBM Research [Melbourne], Benaroya Research Institute [Seattle] (BRI), Okinawa Institute of Science and Technology Graduate University, Keio University, Department of Computing and Mathematical sciences, members, SBML Level 3 Community, Université de Lausanne (UNIL), Università degli Studi di Milano-Bicocca [Milano] (UNIMIB), Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), University of California, Universiteit Leiden [Leiden], Centre National de la Recherche Scientifique (CNRS)-Université Joseph Fourier - Grenoble 1 (UJF)-Centre National de la Recherche Scientifique (CNRS)-Université Joseph Fourier - Grenoble 1 (UJF)-Inria Grenoble - Rhône-Alpes, Humboldt University of Berlin, Universität Heidelberg [Heidelberg], Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Humboldt-Universität zu Berlin, University of California-University of California, Université de Lausanne (UNIL)-Université de Lausanne (UNIL), Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Département de Biologie - ENS Paris, École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM), University of Oxford [Oxford], University of California [Irvine] (UCI), Biotechnology and Biological Sciences Research Council, Computer Science, Institut de biologie de l'ENS Paris (UMR 8197/1024) (IBENS), and Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Paris (ENS Paris)
- Subjects
computational modeling ,Medicine (General) ,Markup language ,[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,INFORMATION ,Interoperability ,interoperability ,Review ,[SDV.BC.BC]Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,ANNOTATION ,0302 clinical medicine ,Software ,file forma ,Models ,Biology (General) ,0303 health sciences ,Computational model ,Applied Mathematics ,Systems Biology ,systems biology ,File format ,3. Good health ,Networking and Information Technology R&D ,Networking and Information Technology R&D (NITRD) ,Computational Theory and Mathematics ,SIMULATION ,General Agricultural and Biological Sciences ,STANDARDS ,REPOSITORY ,Information Systems ,QH301-705.5 ,Bioinformatics ,Systems biology ,Software ecosystem ,Reviews ,Bioengineering ,Methods & Resources ,Biology ,MARKUP LANGUAGE ,Models, Biological ,SBML Level 3 Community members ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,R5-920 ,Animals ,Humans ,SBML ,reproducibility ,030304 developmental biology ,ENVIRONMENT ,General Immunology and Microbiology ,file format ,business.industry ,Computational Biology ,Biological ,ONTOLOGY ,Metabolism ,Logistic Models ,Biochemistry and Cell Biology ,Other Biological Sciences ,Software engineering ,business ,030217 neurology & neurosurgery - Abstract
Systems biology has experienced dramatic growth in the number, size, and complexity of computational models. To reproduce simulation results and reuse models, researchers must exchange unambiguous model descriptions. We review the latest edition of the Systems Biology Markup Language (SBML), a format designed for this purpose. A community of modelers and software authors developed SBML Level 3 over the past decade. Its modular form consists of a core suited to representing reaction‐based models and packages that extend the core with features suited to other model types including constraint‐based models, reaction‐diffusion models, logical network models, and rule‐based models. The format leverages two decades of SBML and a rich software ecosystem that transformed how systems biologists build and interact with models. More recently, the rise of multiscale models of whole cells and organs, and new data sources such as single‐cell measurements and live imaging, has precipitated new ways of integrating data with models. We provide our perspectives on the challenges presented by these developments and how SBML Level 3 provides the foundation needed to support this evolution., Over the past two decades, scientists from different fields have been developing SBML, a standard format for encoding computational models in biology and medicine. This article summarizes recent progress and gives perspectives on emerging challenges.
- Published
- 2020
- Full Text
- View/download PDF
43. Comprehensive Transcriptome Profiling of Cryptic
- Author
-
Jenny L, Smith, Rhonda E, Ries, Tiffany, Hylkema, Todd A, Alonzo, Robert B, Gerbing, Marianne T, Santaguida, Lisa, Eidenschink Brodersen, Laura, Pardo, Carrie L, Cummings, Keith R, Loeb, Quy, Le, Suzan, Imren, Amanda R, Leonti, Alan S, Gamis, Richard, Aplenc, E Anders, Kolb, Jason E, Farrar, Timothy J, Triche, Cu, Nguyen, Daoud, Meerzaman, Michael R, Loken, Vivian G, Oehler, Hamid, Bolouri, and Soheil, Meshinchi
- Subjects
Adult ,Male ,Oncogene Proteins, Fusion ,Gene Expression Profiling ,Infant, Newborn ,Infant ,Middle Aged ,Prognosis ,Receptors, GABA-A ,CD56 Antigen ,Article ,Leukemia, Myeloid, Acute ,MicroRNAs ,Young Adult ,Child, Preschool ,Mutation ,Biomarkers, Tumor ,Humans ,Female ,RNA, Messenger ,Follow-Up Studies - Abstract
PURPOSE: A cryptic inv(16)(p13.3q24.3) encoding the CBFA2T3-GLIS2 fusion is associated with poor outcome in infants with acute megakaryocytic leukemia. We aimed to broaden our understanding of the pathogenesis of this fusion through transcriptome profiling. EXPERIMENTAL DESIGN: Available RNA from children and young adults with de novo AML (N=1,049) underwent transcriptome sequencing (mRNA and miRNA). Transcriptome profiles for those with the CBFA2T3-GLIS2 fusion (N=24) and without (N=1,025) were contrasted to define fusion-specific miRNAs, genes, and pathways. Clinical annotations defined distinct fusion-associated disease characteristics and outcomes. RESULTS: The CBFA2T3-GLIS2 fusion was restricted to infants < 3 years-old (p
- Published
- 2019
44. Neutrophil content predicts lymphocyte depletion and anti-PD1 treatment failure in NSCLC
- Author
-
A. McGarry Houghton, Melissa Shipley, Jill McKay-Fleisch, Afshin Mashadi-Hossein, Gavin Meredith, Julia Kargl, John A. Zebala, Robert H. Pierce, Grace H. Y. Yang, Jeffrey C. Thompson, Dean Y. Maeda, Huajia Zhang, Steven M. Albelda, Xiaodong Zhu, Christina S. Baik, Hamid Bolouri, Mary W. Redman, and Travis J. Friesen
- Subjects
0301 basic medicine ,Male ,Cell type ,Myeloid ,Lung Neoplasms ,Neutrophils ,medicine.medical_treatment ,T cell ,Programmed Cell Death 1 Receptor ,Datasets as Topic ,CD8-Positive T-Lymphocytes ,Receptors, Interleukin-8B ,Flow cytometry ,Receptors, Interleukin-8A ,Cohort Studies ,03 medical and health sciences ,Leukocyte Count ,Mice ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Lymphocytes, Tumor-Infiltrating ,Carcinoma, Non-Small-Cell Lung ,medicine ,Animals ,Humans ,Treatment Failure ,Aged ,medicine.diagnostic_test ,business.industry ,Gene Expression Profiling ,General Medicine ,Immunotherapy ,Middle Aged ,medicine.disease ,Flow Cytometry ,Immunohistochemistry ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Female ,business ,CD8 ,Progressive disease ,Research Article - Abstract
Immune checkpoint inhibitor (ICI) treatment has recently become a first-line therapy for many non–small cell lung cancer (NSCLC) patients. Unfortunately, most NSCLC patients are refractory to ICI monotherapy, and initial attempts to address this issue with secondary therapeutics have proven unsuccessful. To identify entities precluding CD8(+) T cell accumulation in this process, we performed unbiased analyses on flow cytometry, gene expression, and multiplexed immunohistochemical data from a NSCLC patient cohort. The results revealed the presence of a myeloid-rich subgroup, which was devoid of CD4(+) and CD8(+) T cells. Of all myeloid cell types assessed, neutrophils were the most highly associated with the myeloid phenotype. Additionally, the ratio of CD8(+) T cells to neutrophils (CD8/PMN) within the tumor mass optimally distinguished between active and myeloid cases. This ratio was also capable of showing the separation of patients responsive to ICI therapy from those with stable or progressive disease in 2 independent cohorts. Tumor-bearing mice treated with a combination of anti-PD1 and SX-682 (CXCR1/2 inhibitor) displayed relocation of lymphocytes from the tumor periphery into a malignant tumor, which was associated with induction of IFN-γ–responsive genes. These results suggest that neutrophil antagonism may represent a viable secondary therapeutic strategy to enhance ICI treatment outcomes.
- Published
- 2019
45. PD62-06 DISTINCT GENOMIC HALLMARKS EXIST BETWEEN METASTATIC UPPER AND LOWER TRACT UROTHELIAL CARCINOMA
- Author
-
Sonali Arora, Lori Kollath, Andrew C. Hsieh, Lisa McFerrin, Michael T. Schweizer, Heather H. Cheng, Funda Vakar-Lopez, Petros Grivas, Hamid Bolouri, Jonathan L. Wright, Brian Winters, Bruce Montgomery, Navonil De Sarkar, Evan Y. Yu, and Hung-Ming Lam
- Subjects
business.industry ,Urology ,Cancer research ,Medicine ,business ,Urothelial carcinoma - Abstract
INTRODUCTION AND OBJECTIVES:Although the genomic landscape of LTUC is well studied, less is known about UTUC, including in the metastatic sites. We evaluated and compared genomic features of metast...
- Published
- 2019
- Full Text
- View/download PDF
46. Integrative network modeling reveals mechanisms underlying T cell exhaustion
- Author
-
Andrew Dervan, Matthew Trotter, Christopher Mark Hill, Brian Fox, Hamid Bolouri, Alexander V. Ratushny, Anne-Renee van der Vuurst de Vries, Joshua Beilke, Pallavur Sivakumar, Paul Shannon, Rebecca Johnson, Mary Young, Lu Huang, Douglas Bassett, and C.C. Santini
- Subjects
0301 basic medicine ,Computer science ,T cell ,Datasets as Topic ,lcsh:Medicine ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Network topology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Computer Simulation ,Enhancer of Zeste Homolog 2 Protein ,Gene Regulatory Networks ,RNA-Seq ,lcsh:Science ,Oligonucleotide Array Sequence Analysis ,Network model ,Multidisciplinary ,Node (networking) ,lcsh:R ,Models, Immunological ,Translational research ,030104 developmental biology ,medicine.anatomical_structure ,Preclinical research ,030220 oncology & carcinogenesis ,lcsh:Q ,Immunologic Memory ,Neuroscience ,Signal Transduction - Abstract
Failure to clear antigens causes CD8+ T cells to become increasingly hypo-functional, a state known as exhaustion. We combined manually extracted information from published literature with gene expression data from diverse model systems to infer a set of molecular regulatory interactions that underpin exhaustion. Topological analysis and simulation modeling of the network suggests CD8+ T cells undergo 2 major transitions in state following stimulation. The time cells spend in the earlier pro-memory/proliferative (PP) state is a fixed and inherent property of the network structure. Transition to the second state is necessary for exhaustion. Combining insights from network topology analysis and simulation modeling, we predict the extent to which each node in our network drives cells towards an exhausted state. We demonstrate the utility of our approach by experimentally testing the prediction that drug-induced interference with EZH2 function increases the proportion of pro-memory/proliferative cells in the early days post-activation.
- Published
- 2019
- Full Text
- View/download PDF
47. Publisher Correction: The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions
- Author
-
Hamid Bolouri, Jason E Farrar, Timothy Triche, Rhonda E Ries, Emilia L Lim, Todd A Alonzo, Yussanne Ma, Richard Moore, Andrew J Mungall, Marco A Marra, Jinghui Zhang, Xiaotu Ma, Yu Liu, Yanling Liu, Jaime M Guidry Auvil, Tanja M Davidsen, Patee Gesuwan, Leandro C Hermida, Bodour Salhia, Stephen Capone, Giridharan Ramsingh, Christian Michel Zwaan, Sanne Noort, Stephen R Piccolo, E Anders Kolb, Alan S Gamis, Malcolm A Smith, Daniela S Gerhard, and Soheil Meshinchi
- Subjects
Chromosome Aberrations ,Leukemia, Myeloid, Acute ,Mutation ,Humans ,General Medicine ,DNA Methylation ,Child ,Transcriptome ,General Biochemistry, Genetics and Molecular Biology ,Article - Abstract
We present the molecular landscape of pediatric acute myeloid leukemia (AML) and characterize nearly 1,000 participants in Children's Oncology Group (COG) AML trials. The COG-National Cancer Institute (NCI) TARGET AML initiative assessed cases by whole-genome, targeted DNA, mRNA and microRNA sequencing and CpG methylation profiling. Validated DNA variants corresponded to diverse, infrequent mutations, with fewer than 40 genes mutated in2% of cases. In contrast, somatic structural variants, including new gene fusions and focal deletions of MBNL1, ZEB2 and ELF1, were disproportionately prevalent in young individuals as compared to adults. Conversely, mutations in DNMT3A and TP53, which were common in adults, were conspicuously absent from virtually all pediatric cases. New mutations in GATA2, FLT3 and CBL and recurrent mutations in MYC-ITD, NRAS, KRAS and WT1 were frequent in pediatric AML. Deletions, mutations and promoter DNA hypermethylation convergently impacted Wnt signaling, Polycomb repression, innate immune cell interactions and a cluster of zinc finger-encoding genes associated with KMT2A rearrangements. These results highlight the need for and facilitate the development of age-tailored targeted therapies for the treatment of pediatric AML.
- Published
- 2019
48. From DNA Sequence to Network Behavior: Functional Properties of Genetic Regulatory Networks.
- Author
-
Hamid Bolouri
- Published
- 2004
- Full Text
- View/download PDF
49. SBML Level 3: an extensible format for the exchange and reuse of biological models
- Author
-
Sarah M, Keating, Dagmar, Waltemath, Matthias, König, Fengkai, Zhang, Andreas, Dräger, Claudine, Chaouiya, Frank T, Bergmann, Andrew, Finney, Colin S, Gillespie, Tomáš, Helikar, Stefan, Hoops, Rahuman S, Malik‐Sheriff, Stuart L, Moodie, Ion I, Moraru, Chris J, Myers, Aurélien, Naldi, Brett G, Olivier, Sven, Sahle, James C, Schaff, Lucian P, Smith, Maciej J, Swat, Denis, Thieffry, Leandro, Watanabe, Darren J, Wilkinson, Michael L, Blinov, Kimberly, Begley, James R, Faeder, Harold F, Gómez, Thomas M, Hamm, Yuichiro, Inagaki, Wolfram, Liebermeister, Allyson L, Lister, Daniel, Lucio, Eric, Mjolsness, Carole J, Proctor, Karthik, Raman, Nicolas, Rodriguez, Clifford A, Shaffer, Bruce E, Shapiro, Joerg, Stelling, Neil, Swainston, Naoki, Tanimura, John, Wagner, Martin, Meier‐Schellersheim, Herbert M, Sauro, Bernhard, Palsson, Hamid, Bolouri, Hiroaki, Kitano, Akira, Funahashi, Henning, Hermjakob, John C, Doyle, Michael, Hucka, Richard R, Adams, Nicholas A, Allen, Bastian R, Angermann, Marco, Antoniotti, Gary D, Bader, Jan, Červený, Mélanie, Courtot, Chris D, Cox, Piero, Dalle Pezze, Emek, Demir, William S, Denney, Harish, Dharuri, Julien, Dorier, Dirk, Drasdo, Ali, Ebrahim, Johannes, Eichner, Johan, Elf, Lukas, Endler, Chris T, Evelo, Christoph, Flamm, Ronan MT, Fleming, Martina, Fröhlich, Mihai, Glont, Emanuel, Gonçalves, Martin, Golebiewski, Hovakim, Grabski, Alex, Gutteridge, Damon, Hachmeister, Leonard A, Harris, Benjamin D, Heavner, Ron, Henkel, William S, Hlavacek, Bin, Hu, Daniel R, Hyduke, Hidde, Jong, Nick, Juty, Peter D, Karp, Jonathan R, Karr, Douglas B, Kell, Roland, Keller, Ilya, Kiselev, Steffen, Klamt, Edda, Klipp, Christian, Knüpfer, Fedor, Kolpakov, Falko, Krause, Martina, Kutmon, Camille, Laibe, Conor, Lawless, Lu, Li, Leslie M, Loew, Rainer, Machne, Yukiko, Matsuoka, Pedro, Mendes, Huaiyu, Mi, Florian, Mittag, Pedro T, Monteiro, Kedar Nath, Natarajan, Poul MF, Nielsen, Tramy, Nguyen, Alida, Palmisano, Jean‐Baptiste, Pettit, Thomas, Pfau, Robert D, Phair, Tomas, Radivoyevitch, Johann M, Rohwer, Oliver A, Ruebenacker, Julio, Saez‐Rodriguez, Martin, Scharm, Henning, Schmidt, Falk, Schreiber, Michael, Schubert, Roman, Schulte, Stuart C, Sealfon, Kieran, Smallbone, Sylvain, Soliman, Melanie I, Stefan, Devin P, Sullivan, Koichi, Takahashi, Bas, Teusink, David, Tolnay, Ibrahim, Vazirabad, Axel, Kamp, Ulrike, Wittig, Clemens, Wrzodek, Finja, Wrzodek, Ioannis, Xenarios, Takahiro G, Yamada, Anna, Zhukova, Jeremy, Zucker, Sarah M, Keating, Dagmar, Waltemath, Matthias, König, Fengkai, Zhang, Andreas, Dräger, Claudine, Chaouiya, Frank T, Bergmann, Andrew, Finney, Colin S, Gillespie, Tomáš, Helikar, Stefan, Hoops, Rahuman S, Malik‐Sheriff, Stuart L, Moodie, Ion I, Moraru, Chris J, Myers, Aurélien, Naldi, Brett G, Olivier, Sven, Sahle, James C, Schaff, Lucian P, Smith, Maciej J, Swat, Denis, Thieffry, Leandro, Watanabe, Darren J, Wilkinson, Michael L, Blinov, Kimberly, Begley, James R, Faeder, Harold F, Gómez, Thomas M, Hamm, Yuichiro, Inagaki, Wolfram, Liebermeister, Allyson L, Lister, Daniel, Lucio, Eric, Mjolsness, Carole J, Proctor, Karthik, Raman, Nicolas, Rodriguez, Clifford A, Shaffer, Bruce E, Shapiro, Joerg, Stelling, Neil, Swainston, Naoki, Tanimura, John, Wagner, Martin, Meier‐Schellersheim, Herbert M, Sauro, Bernhard, Palsson, Hamid, Bolouri, Hiroaki, Kitano, Akira, Funahashi, Henning, Hermjakob, John C, Doyle, Michael, Hucka, Richard R, Adams, Nicholas A, Allen, Bastian R, Angermann, Marco, Antoniotti, Gary D, Bader, Jan, Červený, Mélanie, Courtot, Chris D, Cox, Piero, Dalle Pezze, Emek, Demir, William S, Denney, Harish, Dharuri, Julien, Dorier, Dirk, Drasdo, Ali, Ebrahim, Johannes, Eichner, Johan, Elf, Lukas, Endler, Chris T, Evelo, Christoph, Flamm, Ronan MT, Fleming, Martina, Fröhlich, Mihai, Glont, Emanuel, Gonçalves, Martin, Golebiewski, Hovakim, Grabski, Alex, Gutteridge, Damon, Hachmeister, Leonard A, Harris, Benjamin D, Heavner, Ron, Henkel, William S, Hlavacek, Bin, Hu, Daniel R, Hyduke, Hidde, Jong, Nick, Juty, Peter D, Karp, Jonathan R, Karr, Douglas B, Kell, Roland, Keller, Ilya, Kiselev, Steffen, Klamt, Edda, Klipp, Christian, Knüpfer, Fedor, Kolpakov, Falko, Krause, Martina, Kutmon, Camille, Laibe, Conor, Lawless, Lu, Li, Leslie M, Loew, Rainer, Machne, Yukiko, Matsuoka, Pedro, Mendes, Huaiyu, Mi, Florian, Mittag, Pedro T, Monteiro, Kedar Nath, Natarajan, Poul MF, Nielsen, Tramy, Nguyen, Alida, Palmisano, Jean‐Baptiste, Pettit, Thomas, Pfau, Robert D, Phair, Tomas, Radivoyevitch, Johann M, Rohwer, Oliver A, Ruebenacker, Julio, Saez‐Rodriguez, Martin, Scharm, Henning, Schmidt, Falk, Schreiber, Michael, Schubert, Roman, Schulte, Stuart C, Sealfon, Kieran, Smallbone, Sylvain, Soliman, Melanie I, Stefan, Devin P, Sullivan, Koichi, Takahashi, Bas, Teusink, David, Tolnay, Ibrahim, Vazirabad, Axel, Kamp, Ulrike, Wittig, Clemens, Wrzodek, Finja, Wrzodek, Ioannis, Xenarios, Takahiro G, Yamada, Anna, Zhukova, and Jeremy, Zucker
- Abstract
Systems biology has experienced dramatic growth in the number, size, and complexity of computational models. To reproduce simulation results and reuse models, researchers must exchange unambiguous model descriptions. We review the latest edition of the Systems Biology Markup Language (SBML), a format designed for this purpose. A community of modelers and software authors developed SBML Level 3 over the past decade. Its modular form consists of a core suited to representing reaction-based models and packages that extend the core with features suited to other model types including constraint-based models, reaction-diffusion models, logical network models, and rule-based models. The format leverages two decades of SBML and a rich software ecosystem that transformed how systems biologists build and interact with models. More recently, the rise of multiscale models of whole cells and organs, and new data sources such as single-cell measurements and live imaging, has precipitated new ways of integrating data with models. We provide our perspectives on the challenges presented by these developments and how SBML Level 3 provides the foundation needed to support this evolution., source:https://www.embopress.org/doi/full/10.15252/msb.20199110
- Published
- 2020
50. miR-155 expression and correlation with clinical outcome in pediatric AML: A report from Children's Oncology Group
- Author
-
Susana C. Raimondi, Michael R. Loken, Maya D. Hughes, Alan S. Gamis, Todd A. Alonzo, Vivian G. Oehler, Hamid Bolouri, Ranjani Ramamurthy, Yi-Cheng Wang, Robert B. Gerbing, Valerie A. Morris, Betsy A. Hirsch, and Soheil Meshinchi
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Myeloid leukemia ,Hematology ,Disease ,Pediatric AML ,Correlation ,miR-155 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cog ,Quartile ,030220 oncology & carcinogenesis ,Internal medicine ,Pediatrics, Perinatology and Child Health ,Cohort ,medicine ,business - Abstract
BACKGROUND Aberrant expression of microRNA-155 (miR-155) has been implicated in acute myeloid leukemia (AML) and associated with clinical outcome. PROCEDURE We evaluated miR-155 expression in 198 children with normal karyotype AML (NK-AML) enrolled in Children's Oncology Group (COG) AML trial AAML0531 and correlated miR-155 expression levels with disease characteristics and clinical outcome. Patients were divided into quartiles (Q1-Q4) based on miR-155 expression level, and disease characteristics were then evaluated and correlated with miR-155 expression. RESULTS MiR-155 expression varied over 4-log10-fold range relative to its expression in normal marrow with a median expression level of 0.825 (range 0.043-25.630) for the entire study cohort. Increasing miR-155 expression was highly associated with the presence of FLT3/ITD mutations (P < 0.001) and high-risk disease (P < 0.001) and inversely associated with standard-risk (P = 0.008) and low-risk disease (P = 0.041). Patients with highest miR-155 expression had a complete remission (CR) rate of 46% compared with 82% in low expressers (P < 0.001) with a correspondingly lower event-free (EFS) and overall survival (OS) (P < 0.001 and P = 0.002, respectively). In a multivariate model that included molecular risk factors, high miR-155 expression remained a significant independent predictor of OS (P = 0.022) and EFS (0.019). CONCLUSIONS High miR-155 expression is an adverse prognostic factor in pediatric NK-AML patients. Specifically, high miR-155 expression not only correlates with FLT3/ITD mutation status and high-risk disease but it is also an independent predictor of worse EFS and OS.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.