FARE2019 WINNERS Sorted by Institute

Total Page:16

File Type:pdf, Size:1020Kb

FARE2019 WINNERS Sorted by Institute FARE2019 WINNERS Sorted By Institute Ling Zhang Postdoctoral Fellow CC Radiology/Imaging/PET and Neuroimaging Convolutional Invasion and Expansion Networks for Tumor Growth Prediction This study investigates the possibility of using deep learning to model and predict tumor growth, which has long been formulated by mathematical modeling. Tumor growth is associated with two fundamental processes: cell invasion and mass-effect. Medical imaging data provides non-invasive and in vivo- measurements of these underlying tumor physiological parameters over time. We propose a two- stream end-to-end convolutional neural network (ConvNet) to directly represent and learn the cell invasion and mass-effect from longitudinal medical images, and to predict the subsequent involvement regions of a tumor. The network architecture can easily be trained on population data and personalized to a target patient, unlike most previous mathematical modeling methods that fail to incorporate population data. Experiments are taken on a longitudinal pancreatic tumor data set with comparison with a state-of-the-art mathematical model-based approach. To the best of our knowledge, this is the first time to use learnable ConvNet models for explicitly capturing the two fundamental processes of tumor growth. The invasion network can make its prediction based on the metabolic rate, cell density and tumor boundary, all derived from the multi-model imaging data. Mass-effect â the mechanical force exerted by the growing tumor â can be approximated by the expansion/shrink motion (magnitude and orientation) of the tumor mass. This expansion/shrink cue is captured by optical flow computing, based on which the expansion network is trained to infer tumor growth. To exploit the inherent correlations among the invasion and expansion information, we study and evaluate three different network architectures, named: early-fusion, late-fusion, and end-to-end fusion. Our proposed ConvNet architectures can be both trained using population data and personalized to a target patient. The proposed ConvNets are tested on a NVIDIA TITAN X Pascal GPU of 12 GB of memory. Quantitative experiments on a pancreatic tumor data set show that the proposed method has a Dice coefficient of 86.8%, and RVD (relative volume difference) of 6.6%, substantially outperforms a state-of-the-art mathematical model-based approach (Dice=84.4%, RVD=13.9%). The proposed ConvNets requires ~ 5 mins for training and personalization, and 15 s for prediction per patient - significantly faster than the state-of-the-art model-based approach (~ 24 hrs â model personalization; 21 s â simulation). __________________________________________________________________________________ Laetitia Gorisse Visiting Fellow CC Biochemistry - General, Proteins, and Lipids IQGAP1 Binds The Axl Receptor Tyrosine Kinase And Inhibits Its Signaling IQGAP1 is a scaffold protein that integrates cell signaling pathways by binding several growth factor receptors and intracellular signaling molecules (Trends Cell Biol 2015; 25:171-184). The Axl tyrosine kinase receptor is important in hematopoiesis, the innate immune response, platelet aggregation, engulfment of apoptotic cells and cell survival (Cold Spring Harb Perspect Biol 2013; 5:a009076). Binding of growth arrest-specific protein 6 (Gas6) to immunoglobulin-like (Ig-like) domains of Axl activates intracellular signaling. However, the mechanism of inactivation of the Axl receptor is poorly understood. Here, we investigate the interaction between IQGAP1 and Axl. Using GST pulldown assays and immunoprecipitation from mammalian cell lysates, we found that IQGAP1 associates with Axl in cells. Analysis with multiple discrete fragments of each protein revealed that the binding is direct and mediated by the IQ domain of IQGAP1 and the Ig-like domains of Axl. We observed by proximity ligation assays that Axl:IQGAP1 complexes are located in the cytoplasm, and this interaction is reduced by 64% when cells are incubated with Gas6. Consistent with these findings, reducing IQGAP1 levels in cells by CRISPR/Cas9 enhanced the ability of Gas6 to stimulate both Axl phosphorylation and activation of Akt. Moreover, reduction of IQGAP1 in cells promoted Gas6-stimulated transcription of matrix metalloproteinase (MMP)-2, MMP-3, MMP-9 and urokinase-type plasminogen activator genes. Finally, we observed by confocal microscopy that IQGAP1 downregulates the heterodimerization of Axl with the epidermal growth factor receptor. In summary, we identify IQGAP1 as a previously undescribed suppressor of Axl. These findings differ from the previously reported functions for IQGAP1 as a scaffold that promotes signaling by growth factor receptors (J Biol Chem 2007; 292:3273-3289; J Biol Chem 2011; 286:29734-29747; J Biol Chem 2011; 286:15010-15021). Our data expand the repertoire of binding partners for both Axl and IQGAP1, and provide additional insight into the regulation of Axl function. __________________________________________________________________________________ Youbao Tang Postdoctoral Fellow CC Radiology/Imaging/PET and Neuroimaging Semi-Automatic RECIST Labeling on CT Scans with Cascaded Convolutional Neural Networks Response evaluation criteria in solid tumors (RECIST) is the standard measurement for tumor extent to evaluate treatment responses in cancer patients. The quality of RECIST annotations will affect the assessment result. To perform RECIST annotations, a radiologist first selects an axial image slice and then he or she measures the diameters of the longest axis and the orthogonal short axis. Using RECIST annotation faces two main challenges: 1) measuring tumor diameters requires a lot of professional knowledge and is time-consuming. 2) RECIST marks are often subjective and prone to inconsistency among different observers. However, consistency is critical in assessing actual lesion growth rates, which directly impacts patient treatment options. To overcome these problems, we propose a RECIST estimation method based on a cascaded convolutional neural network. Specifically, the stacked hourglass networks (SHN) is employed for RECIST estimation, where a relationship constraint loss is introduced to improve the estimation accuracy. Regardless of class, the lesion regions may have large variability in sizes, locations and orientations in different images. To make our method robust to these variations, the lesion region first needs to be normalized before feeding into SHN. The spatial transformer network is used for lesion region normalization, where a localization network is designed for lesion region and transformation parameter prediction. Thus, given a region of interest cropped using a bounding box roughly drawn by a radiologist, our method directly outputs RECIST annotations. As a result, our method is semi-automatic, drastically reducing annotation time while keeping the âhuman in the loopâ. In addition, our method can be readily made fully automatic as it can be trivially connected with any lesion localization framework. We train our system on a large scale lesion dataset where RECIST annotations are performed by multiple radiologists over a multi-year period. Experimental results compare our method to the multi-rater annotations in that dataset, plus annotations from two additional radiologists. The mean and standard deviation of inter-reader variation of long diameter are 3.40mm and 5.24mm, while those for the variation between our system and the manual annotations are 2.67mm and 3.95mm, which demonstrate that our system performs more stably and with less variability, suggesting that RECIST annotations can be reliably obtained with reduced labor and time. __________________________________________________________________________________ Kirill Gorshkov Postdoctoral Fellow NCATS Clinical and Translational Research, and Gene Therapy A novel high-throughput assay for Niemann-Pick disease type A small molecule therapeutics discovery Niemann-Pick disease type A (NPA) is a rare and fatal lysosomal storage disease caused by a deficiency of acid sphingomyelinase (ASM) that results in lysosomal sphingomyelin (SM) accumulation. There is no treatment available for NPA. In this study, we have developed a washless, 384-well plate, quantitative high-throughput screening (qHTS) fluorescence assay to identify compounds that alleviate the SM accumulation phenotype by restoring ASM function or other mechanisms. We first loaded NPA patient fibroblasts with green-fluorescent BODIPY-FL-labeled sphingomyelin, followed by treatment with drug compounds at 37 degrees C for 72 hours. The red-fluorescent LysoTracker dye and DRAQ5 nuclear dye were added at the end of the experiment one hour prior to fixation with Mirskyâs fixative, enabling the detection of individual cells using the Mirrorball fluorescence cytometer, which uses fluorometric microvolume assay technology to exclude background signal. The cellular imaging data consisting of cell count and fluorescent intensity measurements were analyzed using the Cellista software. Our primary and follow-up compound screen used wild-type fibroblasts as a control cell line. DMSO and delta tocopherol were used as negative and positive controls, respectively. We first screened the LOPAC drug library consisting of 1280 bioactive compounds producing 19 positive hits. The hit selection criteria consisted of greater than 30% reduction green fluorescence (decreased SM accumulation) without a decrease in cell counts. Seven hits clustered into two distinct classes targeting Receptor
Recommended publications
  • Nucleoporin 107, 62 and 153 Mediate Kcnq1ot1 Imprinted Domain Regulation in Extraembryonic Endoderm Stem Cells
    ARTICLE DOI: 10.1038/s41467-018-05208-2 OPEN Nucleoporin 107, 62 and 153 mediate Kcnq1ot1 imprinted domain regulation in extraembryonic endoderm stem cells Saqib S. Sachani 1,2,3,4, Lauren S. Landschoot1,2, Liyue Zhang1,2, Carlee R. White1,2, William A. MacDonald3,4, Michael C. Golding 5 & Mellissa R.W. Mann 3,4 1234567890():,; Genomic imprinting is a phenomenon that restricts transcription to predominantly one par- ental allele. How this transcriptional duality is regulated is poorly understood. Here we perform an RNA interference screen for epigenetic factors involved in paternal allelic silen- cing at the Kcnq1ot1 imprinted domain in mouse extraembryonic endoderm stem cells. Multiple factors are identified, including nucleoporin 107 (NUP107). To determine NUP107’s role and specificity in Kcnq1ot1 imprinted domain regulation, we deplete Nup107, as well as Nup62, Nup98/96 and Nup153. Nup107, Nup62 and Nup153, but not Nup98/96 depletion, reduce Kcnq1ot1 noncoding RNA volume, displace the Kcnq1ot1 domain from the nuclear periphery, reactivate a subset of normally silent paternal alleles in the domain, alter histone modifications with concomitant changes in KMT2A, EZH2 and EHMT2 occupancy, as well as reduce cohesin interactions at the Kcnq1ot1 imprinting control region. Our results establish an important role for specific nucleoporins in mediating Kcnq1ot1 imprinted domain regulation. 1 Departments of Obstetrics & Gynaecology, and Biochemistry, Western University, Schulich School of Medicine and Dentistry, London, ON N6A 5W9, Canada. 2 Children’s Health Research Institute, London, ON N6C 2V5, Canada. 3 Departments of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA. 4 Magee-Womens Research Institute, Pittsburgh, PA 15213, USA.
    [Show full text]
  • Analysis of Trans Esnps Infers Regulatory Network Architecture
    Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2014 © 2014 Anat Kreimer All rights reserved ABSTRACT Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer eSNPs are genetic variants associated with transcript expression levels. The characteristics of such variants highlight their importance and present a unique opportunity for studying gene regulation. eSNPs affect most genes and their cell type specificity can shed light on different processes that are activated in each cell. They can identify functional variants by connecting SNPs that are implicated in disease to a molecular mechanism. Examining eSNPs that are associated with distal genes can provide insights regarding the inference of regulatory networks but also presents challenges due to the high statistical burden of multiple testing. Such association studies allow: simultaneous investigation of many gene expression phenotypes without assuming any prior knowledge and identification of unknown regulators of gene expression while uncovering directionality. This thesis will focus on such distal eSNPs to map regulatory interactions between different loci and expose the architecture of the regulatory network defined by such interactions. We develop novel computational approaches and apply them to genetics-genomics data in human. We go beyond pairwise interactions to define network motifs, including regulatory modules and bi-fan structures, showing them to be prevalent in real data and exposing distinct attributes of such arrangements. We project eSNP associations onto a protein-protein interaction network to expose topological properties of eSNPs and their targets and highlight different modes of distal regulation.
    [Show full text]
  • Title a New Centrosomal Protein Regulates Neurogenesis By
    Title A new centrosomal protein regulates neurogenesis by microtubule organization Authors: Germán Camargo Ortega1-3†, Sven Falk1,2†, Pia A. Johansson1,2†, Elise Peyre4, Sanjeeb Kumar Sahu5, Loïc Broic4, Camino De Juan Romero6, Kalina Draganova1,2, Stanislav Vinopal7, Kaviya Chinnappa1‡, Anna Gavranovic1, Tugay Karakaya1, Juliane Merl-Pham8, Arie Geerlof9, Regina Feederle10,11, Wei Shao12,13, Song-Hai Shi12,13, Stefanie M. Hauck8, Frank Bradke7, Victor Borrell6, Vijay K. Tiwari§, Wieland B. Huttner14, Michaela Wilsch- Bräuninger14, Laurent Nguyen4 and Magdalena Götz1,2,11* Affiliations: 1. Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany. 2. Physiological Genomics, Biomedical Center, Ludwig-Maximilian University Munich, Germany. 3. Graduate School of Systemic Neurosciences, Biocenter, Ludwig-Maximilian University Munich, Germany. 4. GIGA-Neurosciences, Molecular regulation of neurogenesis, University of Liège, Belgium 5. Institute of Molecular Biology (IMB), Mainz, Germany. 6. Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d’Alacant, Spain. 7. Laboratory for Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany. 8. Research Unit Protein Science, Helmholtz Centre Munich, German Research Center for Environmental Health, Munich, Germany. 9. Protein Expression and Purification Facility, Institute of Structural Biology, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany. 10. Institute for Diabetes and Obesity, Monoclonal Antibody Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany. 11. SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, Ludwig- Maximilian University Munich, Germany. 12. Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, USA 13.
    [Show full text]
  • Cell Type–Dependent Effects of Polo-Like Kinase 1 Inhibition Compared with Targeted Polo Box Interference in Cancer Cell Lines
    3189 Cell type–dependent effects of Polo-like kinase 1 inhibition compared with targeted polo box interference in cancer cell lines Jenny Fink, Karl Sanders, Alexandra Rippl, certain cell lines but highly contrasting effects in others. Sylvia Finkernagel, Thomas L. Beckers, This may point to subtle differences in the molecular and Mathias Schmidt machinery of mitosis regulation in cancer cells. [Mol Cancer Ther 2007;6(12):3189–97] Nycomed GmbH, Konstanz, Germany Introduction Abstract Polo-like kinase 1 (Plk1) has been identified as key player Multiple critical roles within mitosis have been assigned for G2-M transition and mitotic progression in both normal to Polo-like kinase 1 (Plk1), making it an attractive and tumor cells (1). Multiple roles have been assigned to candidate for mitotic targeting of cancer cells. Plk1 Plk1 at the entry into M phase, mitotic spindle formation, contains two domains amenable for targeted interference: condensation and separation of chromosomes, exit from a kinase domain responsible for the enzymatic function mitosis by activation of the anaphase-promoting complex, and a polo box domain necessary for substrate recogni- and in cytokinesis (reviewed in ref. 2). Moreover, recent tion and subcellular localization. Here, we compare two reports implicated an involvement of Plk1 in the resump- approaches for targeted interference with Plk1 function, tion of cell cycle reentry after checkpoint activation through either by a Plk1 small-molecule enzyme inhibitor or by DNA-damaging agents (3). It is therefore not surprising inducible overexpression of the polo box in human cancer that targeted interference with Plk1, primarily by anti- cell lines.
    [Show full text]
  • Low Basal Expression and Slow Induction of IFITM3 Puts Immune
    bioRxiv preprint doi: https://doi.org/10.1101/2019.12.20.885590; this version posted December 23, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 TITLE: Low basal expression and slow induction of IFITM3 puts immune 2 cells at risk of influenza A infection 3 4 Running head (40 characters): 5 Cells at risk of Influenza A infection 6 7 Authors: 8 Dannielle Wellington1,2,*, , Zixi Yin1,2, Liwei Zhang1, Jessica Forbester1,3, Kerry 9 Kite1, Henry Laurenson-Schafer1, Shokouh Makvandi-Nejad1 , Boquan Jin4, 10 Emma Bowes5, Krishnageetha Manoharan5, David Maldonado-Perez5, Clare 11 Verill5, Ian Humphreys3 & Tao Dong1,2,* 12 1. MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe 13 Department of Medicine, Oxford University, Oxford OX3 9DS, UK 14 2. Chinese Academy of Medical Sciences (CAMS) Oxford Institute, Nuffield Department of 15 Medicine, Oxford University, OX3 7BN, UK 16 3. Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff 17 University, Cardiff, CF14 4XN, UK 18 4. Fourth Military Medical University, Xian, China 19 5. Oxford Radcliffe BioBank, Nuffield Department of Surgical Sciences, Oxford University, 20 Oxford OX3 9DU, UK 21 *Corresponding to Dannielle Wellington [email protected] or Tao Dong, 22 [email protected] 23 24 Acknowledgements: 25 The authors would like to thank the following facilities and individuals within 26 the MRC Weatherall Institute (Oxford) for their invaluaBle help in the following 27 experiments: Mass Cytometry Facility and Alain Townsend for kindly providing 28 virus stocks.
    [Show full text]
  • DNA·RNA Triple Helix Formation Can Function As a Cis-Acting Regulatory
    DNA·RNA triple helix formation can function as a cis-acting regulatory mechanism at the human β-globin locus Zhuo Zhoua, Keith E. Gilesa,b,c, and Gary Felsenfelda,1 aLaboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892; bUniversity of Alabama at Birmingham Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL 35294; and cDepartment of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294 Contributed by Gary Felsenfeld, February 4, 2019 (sent for review January 4, 2019; reviewed by James Douglas Engel and Sergei M. Mirkin) We have identified regulatory mechanisms in which an RNA tran- of the criteria necessary to establish the presence of a triplex script forms a DNA duplex·RNA triple helix with a gene or one of its structure, we first describe and characterize triplex formation at regulatory elements, suggesting potential auto-regulatory mecha- the FAU gene in human erythroid K562 cells. FAU encodes a nisms in vivo. We describe an interaction at the human β-globin protein that is a fusion containing fubi, a ubiquitin-like protein, locus, in which an RNA segment embedded in the second intron of and ribosomal protein S30. Although fubi function is unknown, the β-globin gene forms a DNA·RNA triplex with the HS2 sequence posttranslational processing produces S30, a component of the within the β-globin locus control region, a major regulator of glo- 40S ribosome. We used this system to refine methods necessary bin expression. We show in human K562 cells that the triplex is to detect triplex formation and to distinguish it from R-loop stable in vivo.
    [Show full text]
  • The Basal Bodies of Chlamydomonas Reinhardtii Susan K
    Dutcher and O’Toole Cilia (2016) 5:18 DOI 10.1186/s13630-016-0039-z Cilia REVIEW Open Access The basal bodies of Chlamydomonas reinhardtii Susan K. Dutcher1* and Eileen T. O’Toole2 Abstract The unicellular green alga, Chlamydomonas reinhardtii, is a biflagellated cell that can swim or glide. C. reinhardtii cells are amenable to genetic, biochemical, proteomic, and microscopic analysis of its basal bodies. The basal bodies contain triplet microtubules and a well-ordered transition zone. Both the mother and daughter basal bodies assemble flagella. Many of the proteins found in other basal body-containing organisms are present in the Chlamydomonas genome, and mutants in these genes affect the assembly of basal bodies. Electron microscopic analysis shows that basal body duplication is site-specific and this may be important for the proper duplication and spatial organization of these organelles. Chlamydomonas is an excellent model for the study of basal bodies as well as the transition zone. Keywords: Site-specific basal body duplication, Cartwheel, Transition zone, Centrin fibers Phylogeny and conservation of proteins Centrin, SPD2/CEP192, Asterless/CEP152; CEP70, The green lineage or Viridiplantae consists of the green delta-tubulin, and epsilon-tubulin. Chlamydomonas has algae, which include Chlamydomonas, the angiosperms homologs of all of these based on sequence conservation (the land plants), and the gymnosperms (conifers, cycads, except PLK4, CEP152, and CEP192. Several lines of evi- ginkgos). They are grouped together because they have dence suggests that CEP152, CEP192, and PLK4 interact chlorophyll a and b and lack phycobiliproteins. The green [20, 52] and their concomitant absence in several organ- algae together with the cycads and ginkgos have basal isms suggests that other mechanisms exist that allow for bodies and cilia, while the angiosperms and conifers have control of duplication [4].
    [Show full text]
  • Review Article Mitotic Kinases and P53 Signaling
    Hindawi Publishing Corporation Biochemistry Research International Volume 2012, Article ID 195903, 14 pages doi:10.1155/2012/195903 Review Article Mitotic Kinases and p53 Signaling Geun-Hyoung Ha1 and Eun-Kyoung Yim Breuer1, 2 1 Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA 2 Department of Molecular Pharmacology and Therapeutics, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA Correspondence should be addressed to Eun-Kyoung Yim Breuer, [email protected] Received 6 April 2012; Accepted 18 May 2012 Academic Editor: Mandi M. Murph Copyright © 2012 G.-H. Ha and E.-K. Y. Breuer. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Mitosis is tightly regulated and any errors in this process often lead to aneuploidy, genomic instability, and tumorigenesis. Deregulation of mitotic kinases is significantly associated with improper cell division and aneuploidy. Because of their importance during mitosis and the relevance to cancer, mitotic kinase signaling has been extensively studied over the past few decades and, as a result, several mitotic kinase inhibitors have been developed. Despite promising preclinical results, targeting mitotic kinases for cancer therapy faces numerous challenges, including safety and patient selection issues. Therefore, there is an urgent need to better understand the molecular mechanisms underlying mitotic kinase signaling and its interactive network. Increasing evidence suggests that tumor suppressor p53 functions at the center of the mitotic kinase signaling network. In response to mitotic spindle damage, multiple mitotic kinases phosphorylate p53 to either activate or deactivate p53-mediated signaling.
    [Show full text]
  • Ldb1 Is Essential for Development of Nkx2.1 Lineage Derived Gabaergic and Cholinergic Neurons in the Telencephalon
    Developmental Biology 385 (2014) 94–106 Contents lists available at ScienceDirect Developmental Biology journal homepage: www.elsevier.com/locate/developmentalbiology Ldb1 is essential for development of Nkx2.1 lineage derived GABAergic and cholinergic neurons in the telencephalon Yangu Zhao a,n,1, Pierre Flandin b,2, Daniel Vogt b, Alexander Blood a, Edit Hermesz a,3, Heiner Westphal a, John L. R. Rubenstein b,nn a Program on Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA b Department of Psychiatry and the Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, San Francisco, CA 94143, USA article info abstract Article history: The progenitor zones of the embryonic mouse ventral telencephalon give rise to GABAergic and cholinergic Received 1 June 2013 neurons. We have shown previously that two LIM-homeodomain (LIM-HD) transcription factors, Lhx6 and Received in revised form Lhx8, that are downstream of Nkx2.1, are critical for the development of telencephalic GABAergic and 8 October 2013 cholinergic neurons. Here we investigate the role of Ldb1, a nuclear protein that binds directly to all LIM-HD Accepted 9 October 2013 factors, in the development of these ventral telencephalon derived neurons. We show that Ldb1 is expressed Available online 21 October 2013 in the Nkx2.1 cell lineage during embryonic development and in mature neurons. Conditional deletion of Ldb1 Keywords: causes defects in the expression of a series of genes in the ventral telencephalon and severe impairment in the Differentiation tangential migration of cortical interneurons from the ventral telencephalon.
    [Show full text]
  • Broad and Thematic Remodeling of the Surface Glycoproteome on Isogenic
    bioRxiv preprint doi: https://doi.org/10.1101/808139; this version posted October 17, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Broad and thematic remodeling of the surface glycoproteome on isogenic cells transformed with driving proliferative oncogenes Kevin K. Leung1,5, Gary M. Wilson2,5, Lisa L. Kirkemo1, Nicholas M. Riley2,4, Joshua J. Coon2,3, James A. Wells1* 1Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, USA Departments of Chemistry2 and Biomolecular Chemistry3, University of Wisconsin- Madison, Madison, WI, 53706, USA 4Present address Department of Chemistry, Stanford University, Stanford, CA, 94305, USA 5These authors contributed equally *To whom correspondence should be addressed bioRxiv preprint doi: https://doi.org/10.1101/808139; this version posted October 17, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract: The cell surface proteome, the surfaceome, is the interface for engaging the extracellular space in normal and cancer cells. Here We apply quantitative proteomics of N-linked glycoproteins to reveal how a collection of some 700 surface proteins is dramatically remodeled in an isogenic breast epithelial cell line stably expressing any of six of the most prominent proliferative oncogenes, including the receptor tyrosine kinases, EGFR and HER2, and downstream signaling partners such as KRAS, BRAF, MEK and AKT.
    [Show full text]
  • A Molecular and Genetic Analysis of Otosclerosis
    A molecular and genetic analysis of otosclerosis Joanna Lauren Ziff Submitted for the degree of PhD University College London January 2014 1 Declaration I, Joanna Ziff, confirm that the work presented in this thesis is my own. Where information has been derived from other sources, I confirm that this has been indicated in the thesis. Where work has been conducted by other members of our laboratory, this has been indicated by an appropriate reference. 2 Abstract Otosclerosis is a common form of conductive hearing loss. It is characterised by abnormal bone remodelling within the otic capsule, leading to formation of sclerotic lesions of the temporal bone. Encroachment of these lesions on to the footplate of the stapes in the middle ear leads to stapes fixation and subsequent conductive hearing loss. The hereditary nature of otosclerosis has long been recognised due to its recurrence within families, but its genetic aetiology is yet to be characterised. Although many familial linkage studies and candidate gene association studies to investigate the genetic nature of otosclerosis have been performed in recent years, progress in identifying disease causing genes has been slow. This is largely due to the highly heterogeneous nature of this condition. The research presented in this thesis examines the molecular and genetic basis of otosclerosis using two next generation sequencing technologies; RNA-sequencing and Whole Exome Sequencing. RNA–sequencing has provided human stapes transcriptomes for healthy and diseased stapes, and in combination with pathway analysis has helped identify genes and molecular processes dysregulated in otosclerotic tissue. Whole Exome Sequencing has been employed to investigate rare variants that segregate with otosclerosis in affected families, and has been followed by a variant filtering strategy, which has prioritised genes found to be dysregulated during RNA-sequencing.
    [Show full text]
  • Receptor-Arrestin Interactions: the GPCR Perspective
    biomolecules Review Receptor-Arrestin Interactions: The GPCR Perspective Mohammad Seyedabadi 1,2 , Mehdi Gharghabi 3, Eugenia V. Gurevich 4 and Vsevolod V. Gurevich 4,* 1 Department of Toxicology & Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 48471-93698, Iran; [email protected] 2 Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 48167-75952, Iran 3 Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; [email protected] 4 Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; [email protected] * Correspondence: [email protected]; Tel.: +1-615-322-7070; Fax: +1-615-343-6532 Abstract: Arrestins are a small family of four proteins in most vertebrates that bind hundreds of different G protein-coupled receptors (GPCRs). Arrestin binding to a GPCR has at least three functions: precluding further receptor coupling to G proteins, facilitating receptor internalization, and initiating distinct arrestin-mediated signaling. The molecular mechanism of arrestin–GPCR interactions has been extensively studied and discussed from the “arrestin perspective”, focusing on the roles of arrestin elements in receptor binding. Here, we discuss this phenomenon from the “receptor perspective”, focusing on the receptor elements involved in arrestin binding and empha- sizing existing gaps in our knowledge that need to be filled. It is vitally important to understand the role of receptor elements in arrestin activation and how the interaction of each of these elements with arrestin contributes to the latter’s transition to the high-affinity binding state. A more precise knowledge of the molecular mechanisms of arrestin activation is needed to enable the construction of arrestin mutants with desired functional characteristics.
    [Show full text]