Towards Understanding Selective Neuronal Vulnerability: Establishing an In-Vitro Model for Strain Selection

Total Page:16

File Type:pdf, Size:1020Kb

Towards Understanding Selective Neuronal Vulnerability: Establishing an In-Vitro Model for Strain Selection Towards understanding selective neuronal vulnerability: Establishing an in-vitro model for strain selection A thesis submitted in partial fulfilment of the requirements for the degree of Doctor of Philosophy from University College London Alexandra Philiastides MRC Prion Unit at UCL Institute of Prion Diseases University College London 2018 1 Declaration I, Alexandra Philiastides, 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. 2 Dedication This thesis is dedicated to the memory of my dearest grandfather Andreas Philiastides and to my beloved grandparents Michalis and Theano Themistokleous. 3 Acknowledgments Firstly, I would like to express my sincere gratitude to my PhD supervisor Dr. Peter- Christian Kloehn, for the continuous support, scientific input, encouragement and patience throughout my PhD. I would like to thank my previous supervisor Dr. Sarah Lloyd, for her great support, guidance and kind advice during the first 18 months of my PhD. A special thank you to Dr. Craig Brown, who introduced me to cell culture and molecular cloning. It was a real privilege for me to share of his exceptional scientific knowledge which was pivotal for the accomplishment of the work presented in this thesis. I would also like to thank Dr. Juan Ribes, for his continuous support, help with data analysis, experimental planning and protocols and for his encouragement when the PhD seemed like an unending struggle. I would like to express my special appreciation and thanks to my secondary supervisor Prof. Parmjit Jat, for his valuable advice on my research as well as on my career. I am also grateful to my departmental graduate tutor Prof. Elizabeth Fisher, for her tremendous support and advice during the difficult times of my PhD. Many thanks are due to Dr. Graham Jackson, for his insightful comments and valuable suggestions which helped me during the write up of this thesis. This work would not have been possible without the invaluable scientific input and patience of Daniel Yip, Christian Schmidt and Parvin Ahmed who carried out the automated Scrapie Cell assays and devoted their time to help me collect data for my PhD thesis. I am grateful to Nunu Arora, Mark Batchelor, Susan Joiner, Jessica Sells, Adam Wenborn, Dr. Emmanuelle Vire, Emma Quarterman and Gary Adamson for the scientific input, encouragement and help throughout the past four years. I am thankful to my friends Luke Dabin, Laura Pulford, Justin Tosh, Lucy Sheytanova, Nora Thoeng, Matt Rickman, Dr. Karen Cleverley, Billy West, Xun Yu Choong, Julia Ravey, Madeleine Reilly and Shaheen Akhtar, with whom I have shared many happy moments and who were always encouraging throughout my PhD. I would also like to thank my colleagues with whom I have shared the PhD office, for the support and the happy memories which I treasure so much: Lucianne Dobson, James Miller, Charlotte Ridler, Prasanth Sivakumar, David Thomas, Grace O’Regan, Karolin Koriath, Caroline 4 Casey, Angelos Armen, Rachele Saccon, Daniel Wright, Francesca De Giorgio, Rhia Ghosh, Marta Benedekova, Claudia Cannavo, Heather Whittaker, Thanos Dimitriadis, Ines Whitworth, Bernie Simone and Emma Jones. I am grateful to the people in the IT and administration office who were always friendly and happy to help: Ryan Peter, Samantha MacLeod, Amir Mayahi, David Ruegg, Alison Church and Oke Avwenagha. This work would not have been possible without the funding provided by John Collinge through the Medical Research Council. I am thankful and grateful beyond words to my best friend Ruchi Kumari for her immense support and love, and for being the most amazing, caring, genuine and loving friend I could ever ask for. Thank you for the love, the patience, the endless deep conversations, the daily coffee breaks and for believing in me and reminding me that things will always be ok in the end. I would not have been able to go through this without you. To my dearest parents, Antony and Stalo, and to my precious brothers Andreas and Michalis, thank you for the continuous support, the endless love and for helping me overcome situations when continuing seemed impossible. To my amazing fiancé, Charlie, thank you for the love, support, encouragement and never-ending patience. 5 Abstract Prion diseases are fatal neurodegenerative diseases that affect humans and animals. Prion strains, conformational variants of misfolded prion proteins, are thought to be associated with distinct clinical and pathological phenotypes. Why prion strains cause damage in particular areas of the brain is poorly understood. Although prions are innocuous to most cell lines, differences in their tropism to mouse-adapted prion strains have been broadly observed. While some cell lines show broad susceptibility to prion strains, others are highly selective, suggesting that susceptibility to a specific prion strain is determined by distinct cellular factors. The neuroblastoma cell line N2aPK1 (PK1) is refractory to the murine prion strain Me7, but highly susceptible to RML. Intracerebral inoculation of mice with Me7 induces hippocampal neuronal loss, whereas RML does not cause degeneration in this brain region. The PME2 subclone, is a PK1-derived subclone with low susceptibility to Me7 and this was used as the parental line to derive highly Me7-susceptible cells. To understand the molecular underpinning of selective neuronal vulnerability and cell tropism of prion strains, respectively, we first undertook a series of successive sub cloning experiments to identify rare PK1 cell clones that are susceptible to Me7. Initially, Me7-susceptible clones were identified at a frequency of only 4x10-3. The percentage of Me7-susceptible cells increased by 6-fold and 20-fold, respectively, and by the third and final round of sub cloning, 63% of cell clones were highly susceptible to Me7. Persistently infected PME2 cell clones deposited disease-associated PrP (PrPd) in perinuclear and extracellular stores. Strikingly, Me7-refractory PK1 cells were found to be highly susceptible to prions derived from homogenates of chronically Me7- infected PME2 cells, suggesting that a single passage in PME2 cells changed the strain properties of brain-adapted Me7. This cell model provides the first evidence for prion strain adaptation in genetically similar cell clones. The identification of genetically similar cell clones that differ in their ability to adapt prion strains lays the foundation for future work to gain insights into the molecular mechanisms that underlie prion strain adaptation. During the second half of my PhD, I worked on a separate project, investigating the role of Fkbp proteins in molecular mechanisms of prion propagation. While the prion protein gene is the major genetic determinant of susceptibility to prion disease, several studies have identified additional modifier genes that also influence susceptibility and modify the disease phenotype. 6 A microarray gene expression study which correlated the level of mRNA expression, in uninfected brains, from 5 inbred lines of mice, with their respective incubation times identified several potential prion modifier genes including Fkbp9. Lower levels of expression of Fkbp9 correlated with shorter incubation times in mice, following prion infection. These findings were validated in vitro where Scrapie Cell Assays (SCA) in Fkbp9 stably knocked down cell lines showed a significant increase prion propagation. The Fkbp9 protein is part of the immunophilin family of proteins which are peptidyl- propyl cis-trans isomerases. Fkbp proteins have been implicated in aspects of neurodegenerative disease, including accelerating α-synuclein fibrillisation and aggregation (primarily Fkbp12 but also Fkbp38, 52 and 65) and inhibiting tau induced tubulin polymerisation (Fkbp52) in vitro. Fkbp52 also reduced Aβ levels in a fly model of Alzheimer’s disease and Fkbp51 was shown to block tau clearance through the proteasome resulting in oligomerisation. The aim of this project was to characterise the functional roles of Fkbp family members in prion propagation. I generated a panel of N2aPK1 cell lines by stable gene silencing of four different Fkbp genes and employed the SCA to test whether Fkbp knock down (KD) influences prion propagation. For each Fkbp gene, four to eight KD cell lines were generated. Three out of four Fkbp4 (Fkbp52) KD cell lines with over 50% KD of mRNA expression levels showed a significant reduction in the number of PrPSc- positive cells, as quantified in the SCA. Additionally, KD of Fkbp8 in PK1 cells led to a significant reduction in the number of PrPSc-positive cells in four out of the five cell lines screened in the SCA. In contrast to these findings, in some cell lines with a significant reduction in mRNA expression levels (>60%) of the target Fkbp gene, there was no corresponding decrease in the number of PrPSc-positive cells. We reasoned that shRNA off-target effects arising when an shRNA downregulates unintended gene targets through partial sequence complementarity, may mask the effect of KD of the gene of interest. To examine whether an independent gene silencing approach for the examined gene targets recapitulates the results of stable gene silencing, siRNAs were used to transiently knock down Fkbp genes in chronically RML-infected PK1 cells (iS7 cells). Surprisingly, none of the siRNAs against the specified Fkbp genes reduced the number of PrPSc-positive iS7 cells. After establishing which Fkbp proteins affect prion propagation in the SCA, we aimed to carry out in vitro studies to understand the molecular mechanisms by which Fkbp proteins influence prion propagation. After optimisation of expression and cloning strategies, I successfully induced the expression of recombinant Fkbp9 and Fkbp52 proteins. The aim was to use the recombinant proteins in cell-free assays to test whether Fkbp proteins affect prion 7 replication and/or modulate the fibrillisation of recombinant PrPC. In vitro assays with recombinant Fkbp proteins were not carried out as the project was terminated shortly after my primary supervisor left the Unit.
Recommended publications
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Joint Gene Network Construction by Single-Cell RNA Sequencing Data
    bioRxiv preprint doi: https://doi.org/10.1101/2021.07.14.452387; this version posted July 14, 2021. 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 4.0 International license. Biometrics xx, 1–24 DOI: pending...... xxxx 2021 Joint Gene Network Construction by Single-Cell RNA Sequencing Data Meichen Dong1, Yiping He2, Yuchao Jiang 1,3, Fei Zou1,3,∗ 1Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, U.S.A. 2Department of Pathology, Duke University, Durham, North Carolina, U.S.A. 3Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, U.S.A. email: [email protected] Summary: In contrast to differential gene expression analysis at single gene level, gene regulatory networks (GRN) analysis depicts complex transcriptomic interactions among genes for better understandings of underlying genetic architectures of human diseases and traits. Recently, single-cell RNA sequencing (scRNA-seq) data has started to be used for constructing GRNs at a much finer resolution than bulk RNA-seq data and microarray data. However, scRNA- seq data are inherently sparse which hinders direct application of the popular Gaussian graphical models (GGMs). Furthermore, most existing approaches for constructing GRNs with scRNA-seq data only consider gene networks under one condition. To better understand GRNs under different but related conditions with single-cell resolution, we propose to construct Joint Gene Networks with scRNA-seq data (JGNsc) using the GGMs framework.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Protein Network Analyses of Pulmonary Endothelial Cells In
    www.nature.com/scientificreports OPEN Protein network analyses of pulmonary endothelial cells in chronic thromboembolic pulmonary hypertension Sarath Babu Nukala1,8,9*, Olga Tura‑Ceide3,4,5,9, Giancarlo Aldini1, Valérie F. E. D. Smolders2,3, Isabel Blanco3,4, Victor I. Peinado3,4, Manuel Castell6, Joan Albert Barber3,4, Alessandra Altomare1, Giovanna Baron1, Marina Carini1, Marta Cascante2,7,9 & Alfonsina D’Amato1,9* Chronic thromboembolic pulmonary hypertension (CTEPH) is a vascular disease characterized by the presence of organized thromboembolic material in pulmonary arteries leading to increased vascular resistance, heart failure and death. Dysfunction of endothelial cells is involved in CTEPH. The present study describes for the frst time the molecular processes underlying endothelial dysfunction in the development of the CTEPH. The advanced analytical approach and the protein network analyses of patient derived CTEPH endothelial cells allowed the quantitation of 3258 proteins. The 673 diferentially regulated proteins were associated with functional and disease protein network modules. The protein network analyses resulted in the characterization of dysregulated pathways associated with endothelial dysfunction, such as mitochondrial dysfunction, oxidative phosphorylation, sirtuin signaling, infammatory response, oxidative stress and fatty acid metabolism related pathways. In addition, the quantifcation of advanced oxidation protein products, total protein carbonyl content, and intracellular reactive oxygen species resulted increased
    [Show full text]
  • HSP90-Incorporating Chaperome Networks As Biosensor for Disease-Related Pathways in Patient- Specific Midbrain Dopamine Neurons
    ARTICLE DOI: 10.1038/s41467-018-06486-6 OPEN HSP90-incorporating chaperome networks as biosensor for disease-related pathways in patient- specific midbrain dopamine neurons Sarah Kishinevsky1,2,3,4, Tai Wang3, Anna Rodina3, Sun Young Chung1,2, Chao Xu3, John Philip5, Tony Taldone3, Suhasini Joshi3, Mary L. Alpaugh3,14, Alexander Bolaender3, Simon Gutbier6, Davinder Sandhu7, Faranak Fattahi1,2, Bastian Zimmer1,2, Smit K. Shah3, Elizabeth Chang5, Carmen Inda3,15, John Koren 3rd3,16, Nathalie G. Saurat1,2, Marcel Leist 6, Steven S. Gross7, Venkatraman E. Seshan8, Christine Klein9, Mark J. Tomishima1,2,10, Hediye Erdjument-Bromage11,12, Thomas A. Neubert 11,12, Ronald C. Henrickson5, 1234567890():,; Gabriela Chiosis3,13 & Lorenz Studer1,2 Environmental and genetic risk factors contribute to Parkinson’s Disease (PD) pathogenesis and the associated midbrain dopamine (mDA) neuron loss. Here, we identify early PD pathogenic events by developing methodology that utilizes recent innovations in human pluripotent stem cells (hPSC) and chemical sensors of HSP90-incorporating chaperome networks. We show that events triggered by PD-related genetic or toxic stimuli alter the neuronal proteome, thereby altering the stress-specific chaperome networks, which produce changes detected by chemical sensors. Through this method we identify STAT3 and NF-κB signaling activation as examples of genetic stress, and phospho-tyrosine hydroxylase (TH) activation as an example of toxic stress-induced pathways in PD neurons. Importantly, pharmacological inhibition of the stress chaperome network reversed abnormal phospho- STAT3 signaling and phospho-TH-related dopamine levels and rescued PD neuron viability. The use of chemical sensors of chaperome networks on hPSC-derived lineages may present a general strategy to identify molecular events associated with neurodegenerative diseases.
    [Show full text]
  • Identifying the Proteins to Which Small-Molecule Probes and Drugs Bind in Cells
    Identifying the proteins to which small-molecule probes and drugs bind in cells Shao-En Onga,1, Monica Schenonea, Adam A. Margolinb, Xiaoyu Lic, Kathy Doa, Mary K. Doudd, D. R. Mania,b, Letian Kuaie, Xiang Wangd, John L. Woodf, Nicola J. Tollidayc, Angela N. Koehlerd, Lisa A. Marcaurellec, Todd R. Golubb, Robert J. Gouldd, Stuart L. Schreiberd,1, and Steven A. Carra,1 aProteomics Platform, bCancer Biology Program, cChemical Biology Platform, dChemical Biology Program, and eStanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA 02142; and fChemistry Department, Colorado State University, Fort Collins, CO 80523 Contributed by Stuart L. Schreiber, January 15, 2009 (sent for review December 21, 2008) Most small-molecule probes and drugs alter cell circuitry by interact- known (the ‘‘target I.D. problem’’). It could provide strong clues to ing with 1 or more proteins. A complete understanding of the the mechanisms used by SMs to achieve their recognized actions interacting proteins and their associated protein complexes, whether and it could suggest potential unrecognized actions. the compounds are discovered by cell-based phenotypic or target- Strategies for ‘‘target identification’’ have been developed that based screens, is extremely rare. Such a capability is expected to be rely on genetic (9), computational (10, 11) and biochemical (12) highly illuminating—providing strong clues to the mechanisms used principles. Although several key molecular targets have been iden- by small-molecules to achieve their recognized actions and suggest- tified through affinity chromatography (13–15), it has not been ing potential unrecognized actions. We describe a powerful method widely applied as a general solution to target identification for a combining quantitative proteomics (SILAC) with affinity enrichment number of reasons.
    [Show full text]
  • Inhibition of the FKBP Family of Peptidyl Prolyl Isomerases Induces Abortive Translocation and Degradation of the Cellular Prion Protein
    Inhibition of the FKBP Family of Peptidyl Prolyl Isomerases Induces Abortive Translocation and Degradation of the Cellular Prion Protein by Maxime Sawicki A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Biochemistry University of Toronto © Copyright by Maxime Sawicki 2015 Inhibition of the FKBP Family of Peptidyl Prolyl Isomerases Induces Abortive Translocation and Degradation of the Cellular Prion Protein Maxime Sawicki Master of Science Department of Biochemistry University of Toronto 2015 Abstract Prion disorders are a class of neurodegenerative diseases that feature a structural change of the prion protein from its cellular form (PrPC) into its scrapie form (PrPSc). As these disorders are currently incurable, there is a crucial need for novel therapeutic agents. Here, the FDA-approved immunosuppressive drug FK506 was shown to cause an attenuation in the endoplasmic reticulum (ER) translocation of PrPC by exacerbating an intrinsic inefficiency of PrP’s ER-targeting signal sequence, effectively causing the proteasomal degradation of PrPC. Furthermore, the depletion of FKBP10 also caused the degradation of PrPC but at a later stage following translocation into the ER. Additionally, novel FK506 analogues with reduced immunosuppressive properties were shown to be as efficacious as FK506 in downregulating PrPC. Finally, both FK506 treatment and FKBP10 depletion were shown to reduce the levels of PrPSc in chronically infected cell models. These findings offer a new insight into the development of treatments against prion disorders. ii Acknowledgments The completion of the present thesis would not have been possible without the help and support of a number of people. First and foremost, I would like to thank my supervisor, Dr David Williams, for his constant guidance and expertise that allowed me to successfully complete my degree, as well as my committee members, Dr John Glover and Dr Gerold Schmitt-Ulms, for their invaluable advice and suggestions over the course of this project.
    [Show full text]
  • Downloaded and the Kinasefrom Domainthe Pubmed of TOR Server As Input at the Templates
    biomolecules Review Peptidylprolyl Isomerases as In Vivo Carriers for Drugs That Target Various Intracellular Entities Andrzej Galat Service d’Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, F-91191 Gif/Yvette, France; [email protected]; Fax: +33-169089137 Academic Editor: John A. Carver Received: 24 August 2017; Accepted: 26 September 2017; Published: 29 September 2017 Abstract: Analyses of sequences and structures of the cyclosporine A (CsA)-binding proteins (cyclophilins) and the immunosuppressive macrolide FK506-binding proteins (FKBPs) have revealed that they exhibit peculiar spatial distributions of charges, their overall hydrophobicity indexes vary within a considerable level whereas their points isoelectric (pIs) are contained from 4 to 11. These two families of peptidylprolyl cis/trans isomerases (PPIases) have several distinct functional attributes such as: (1) high affinity binding to some pharmacologically-useful hydrophobic macrocyclic drugs; (2) diversified binding epitopes to proteins that may induce transient manifolds with altered flexibility and functional fitness; and (3) electrostatic interactions between positively charged segments of PPIases and negatively charged intracellular entities that support their spatial integration. These three attributes enhance binding of PPIase/pharmacophore complexes to diverse intracellular entities, some of which perturb signalization pathways causing immunosuppression and other system-altering phenomena in humans. Keywords: PPIase; FKBP; cyclophilin; rapamycin; FK506 1. Introduction About forty years ago, two different types of macrocyclic molecules were isolated and shown to possess immunosuppressive activities, such as the cyclic peptide containing non-standard amino acid residues (AAs) cyclosporine A (CsA) and its homologues [1], and two polyketides having L-pipecolic acid ring, namely immunosuppressive macrolide FK506 [2] and rapamycin [3,4].
    [Show full text]
  • Candidate Genes for Alcohol Preference Identified by Expression
    Liang et al. Genome Biology 2010, 11:R11 http://genomebiology.com/2010/11/2/R11 RESEARCH Open Access Candidate genes for alcohol preference identified by expression profiling in alcohol-preferring and -nonpreferring reciprocal congenic rats Tiebing Liang1*, Mark W Kimpel2, Jeanette N McClintick3, Ashley R Skillman1, Kevin McCall4, Howard J Edenberg3, Lucinda G Carr1 Abstract Background: Selectively bred alcohol-preferring (P) and alcohol-nonpreferring (NP) rats differ greatly in alcohol preference, in part due to a highly significant quantitative trait locus (QTL) on chromosome 4. Alcohol consumption scores of reciprocal chromosome 4 congenic strains NP.P and P.NP correlated with the introgressed interval. The goal of this study was to identify candidate genes that may influence alcohol consumption by comparing gene expression in five brain regions of alcohol-naïve inbred alcohol-preferring and P.NP congenic rats: amygdala, nucleus accumbens, hippocampus, caudate putamen, and frontal cortex. Results: Within the QTL region, 104 cis-regulated probe sets were differentially expressed in more than one region, and an additional 53 were differentially expressed in a single region. Fewer trans-regulated probe sets were detected, and most differed in only one region. Analysis of the average expression values across the 5 brain regions yielded 141 differentially expressed cis-regulated probe sets and 206 trans-regulated probe sets. Comparing the present results from inbred alcohol-preferring vs. congenic P.NP rats to earlier results from the reciprocal congenic NP.P vs. inbred alcohol-nonpreferring rats demonstrated that 74 cis-regulated probe sets were differentially expressed in the same direction and with a consistent magnitude of difference in at least one brain region.
    [Show full text]
  • WO 2019/046815 Al 07 March 2019 (07.03.2019) W 1P O PCT
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization I International Bureau (10) International Publication Number (43) International Publication Date WO 2019/046815 Al 07 March 2019 (07.03.2019) W 1P O PCT (51) International Patent Classification: OSTERTAG, Eric [US/US]; 4242 Campus Point Court, C12N 15/90 (2006.01) Suite 700, San Diego, California 82121 (US). RICHTER, Maximilian [US/US]; 473 1Kansas Street, San Diego, Cal¬ (21) International Application Number: ifornia 921 16 (US). CRANERT, Stacey Ann [US/US]; PCT/US20 18/049257 7693 Palmilla Dr. Apt. 2103, San Diego, California 92122 (22) International Filing Date: (US). 31 August 2018 (3 1.08.2018) (74) Agent: MILLER, Katherine J. et al.; COOLEY LLP, (25) Filing Language: English 1299 Pennsylvania Avenue, NW, Suite 700, Washington, District of Columbia 20004 (US). (26) Publication Language: English (81) Designated States (unless otherwise indicated, for every (30) Priority Data: kind of national protection available): AE, AG, AL, AM, 62/552,861 31 August 2017 (3 1.08.2017) US AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, BZ, 62/558,286 13 September 2017 (13.09.2017) US CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, DO, 62/608,546 20 December 2017 (20. 12.2017) US DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, (71) Applicant: POSEIDA THERAPEUTICS, INC. HR, HU, ID, IL, IN, IR, IS, JO, JP, KE, KG, KH, KN, KP, [US/US]; 4242 Campus Point Court, Suite 700, San Diego, KR, KW, KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, California 92121 (US).
    [Show full text]
  • The Human Fk506binding Proteins: Characterization of Human FKBP19
    The human FK506-binding proteins: characterization of human FKBP19 Article (Accepted Version) Rulten, Stuart L, Kinloch, Ross A, Tateossian, Hilda, Robinson, Colin, Gettins, Lucy and Kay, John E (2006) The human FK506-binding proteins: characterization of human FKBP19. Mammalian Genome, 17 (4). pp. 322-331. ISSN 0938-8990 This version is available from Sussex Research Online: http://sro.sussex.ac.uk/id/eprint/22944/ This document is made available in accordance with publisher policies and may differ from the published version or from the version of record. If you wish to cite this item you are advised to consult the publisher’s version. Please see the URL above for details on accessing the published version. Copyright and reuse: Sussex Research Online is a digital repository of the research output of the University. Copyright and all moral rights to the version of the paper presented here belong to the individual author(s) and/or other copyright owners. To the extent reasonable and practicable, the material made available in SRO has been checked for eligibility before being made available. Copies of full text items generally can be reproduced, displayed or performed and given to third parties in any format or medium for personal research or study, educational, or not-for-profit purposes without prior permission or charge, provided that the authors, title and full bibliographic details are credited, a hyperlink and/or URL is given for the original metadata page and the content is not changed in any way. http://sro.sussex.ac.uk The Human FK506-Binding Proteins: Characterisation of Human FKBP19 Stuart L Rulten1*, Ross A Kinloch2, Hilda Tateossian3, Colin Robinson2, Lucy Gettins2 and John E Kay4.
    [Show full text]
  • Adipocyte Protein Carbonylation and Oxidative Stress in Obesity-Linked Mitochondrial Dysfunction and Insulin Resistance
    ADIPOCYTE PROTEIN CARBONYLATION AND OXIDATIVE STRESS IN OBESITY-LINKED MITOCHONDRIAL DYSFUNCTION AND INSULIN RESISTANCE A DISSERTATION SUBMITTED TO THE FACULTY OF THE GRADUATE SCHOOL OF THE UNIVERSITY OF MINNESOTA BY JESSICA MARIE CURTIS IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DR. DAVID A BERNLOHR, ADVISOR SEPTEMBER 2011 © JESSICA MARIE CURTIS 2011 ACKNOWLEDGEMENTS I would like to thank my graduate advisor Dr. Bernlohr for his guidance and support. His positive attitude and patience make him a remarkable teacher, and he has played a pivotal role in my development as a scientist. I especially would like to thank the members of my committee, Drs. Alex Lange, Howard Towle, Edgar Arriaga and Doug Mashek, for their guidance during the last 5 years. Their metabolic expertise and critical reviews of my research has been incredibly helpful. To the members of the Bernlohr Lab, past and present, thank you for the camaraderie and help through the years. You have been an essential part of my graduate experience, creating an environment that is friendly and rewarding of hard work. I must thank Paul Grimsrud for developing the initial GSTA4 studies and for his mentorship. Wendy Hahn has been an essential mentor and GSTA4 project collaborator as well as a great friend. Thank you also to Ann Hertzel for your critical eye and always knowing where everything is; Tina Hellberg for your amazing attention to detail; Rocio Foncea for your fun spirit and antioxidant knowledge; Marissa Lee, the caretaker of cells. Past lab members who shaped my graduated career include Anne Smith, Brian Thomspon, Brian Wiczer, Sandra Lobo, Jason Fowler, and honorary lab member Sri Bandhakavi.
    [Show full text]