Long-Lasting Memory Impairments After Subchronic Immune Challenge

Total Page:16

File Type:pdf, Size:1020Kb

Load more

Long-Lasting Memory Impairments after Subchronic Immune Challenge by Daria Tchessalova A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Neuroscience) in the University of Michigan 2019 Doctoral Committee: Assistant Professor Natalie C. Tronson, Chair Professor Anuska Andjekovic-Zochowska Professor Gabriel Corfas Assistant Professor Jacek Debiec Professor Shigeki Iwase Daria Tchessalova [email protected] ORCID iD: 0000-0001-8619-0922 © Daria Tchessalova 2019 Acknowledgements As a graduate student at the University of Michigan, I have had the exceptional privilege of training in a research-intensive, collaborative, supportive academic environment. I would like to sincerely thank my dissertation mentor, Dr. Natalie Tronson for her close mentorship, endless efforts in guiding me to expand my critical thinking and problem solving skills, encouragement in presenting my graduate work at conferences and presentations, and all of her support in my intellectual growth. Dr. Tronson has taught me to exercise the conceptual framework necessary for effective presentations, grant proposals, and manuscripts. I very much appreciate all of her efforts to help me become the scientist and researcher that I am today. I admire her for her effortless ability to efficiently achieve long-term project goals and to assist students with any issues, even last minute issues, related to their research projects or personal lives. I would like to thank the members of my committee for all of their wonderful suggestions regarding my research project throughout the years. Dr. Jacek Debiec has been a very helpful and encouraging mentor. His philosophical approach to science inspires me to think beyond the scope of specific research questions in my project to better understand how my research impacts individuals suffering from various neurological disorders and diseases. I would like to sincerely thank Dr. Gabriel Corfas for allowing me to explore research questions that I was most passionate about in the field of neuron-glia interactions as a rotating student. While I had not ii received the results that I had hoped for, I learned to effectively design experiments, troubleshoot, and, eventually, avoid letting my stubbornness get in the way of my progress. I would also like to thank Dr. Corfas for his extremely helpful feedback on my dissertation project for my 4th year talk. I would like to sincerely thank our collaborators, Dr. Anuska Andjelkovic- Zochowska and Dr. Svetlana Stamatovic for all of their wonderful guidance and their help with the blood-brain barrier studies as well as for all of the interesting discussions regarding aging and Alzheimer’s disease. I have very much enjoyed our collaboration on cognitive dysfunction in aging and the role of Sirt1. I would also like to very much thank Dr. Shigeki Iwase, Christina Vallianatos, and Tricia Garay for their advice, support, and assistance with prepping of the RNA samples. We would very much like to thank them for allowing us to use their laboratory equipment for RNA extraction. I would also like to thank Dr. Iwase for all of his mentorship and advice for organizing the 2018 NGP Spring Symposium on Neuroepigenetics. Drs. Richard McEachin, Rebecca Tagett, and Robert Lyons from the Bioinformatics for at the University of Michigan have been a huge asset to our gene expression studies. They have advised us regarding the best sequencing method for the RNA-sequencing experiments and performed the Bioinformatics analysis for differentially expressed genes. I would also like to thank Dave Bridges for performing the TRANSFAC analysis in R for predicted transcription factors for our differently expressed genes set. Throughout my time at the University of Michigan, I have been very fortunate to get to know some of the nicest and most helpful co-workers I have ever met. I would like to thank Dr. Ashley Keiser for all her help with my research project over the years as well as her great jokes and supportive nature with any challenges that lay ahead. I would like to very much thank Lacie iii Turnbull for her endless assistance, support, and caring nature that very much helped me get oriented when I first started in the lab. Dr. Katie Collette was also one of the most supportive and caring research and writing mentors and life coaches I have met. I would like to thank Caitlin Posillico for all of her helpful guidance and wonderful conversations related to our research projects and beyond as well as her kind introductions to various members of the Psychoneuroimmunology Research Society. While only in the lab for a short time, Melanie Gil has been an amazing emotional and physical support system with a great passion and can do attitude that has helped me to carry on, even during the toughest days. Brynne Raines has been a very helpful and dedicated lab manager. I would like to thank the undergraduate students who have provided excellent assistance with my research project, especially Grayson Buning who performed behavioral scoring of depressive-like behaviors, and imaging, and cell counting for c-Fos analysis, Rachel Fenberg who completed hand-scoring analysis of various behavioral tests, as well as Faith Best who helped me with analysis of behavioral testing, blood-brain barrier permeability, and microglial activation experiments. Additionally, I would like to thank Eugene Shim for his positive attitude and entertaining nature. iv TABLE OF CONTENTS ACKNOWLEDGEMENTS ............................................................................................. ii TABLE OF CONTENTS ..................................................................................................v LIST OF TABLES .......................................................................................................... vii LIST OF FIGURES ....................................................................................................... viii ABSTRACT ...................................................................................................................... xi Chapter I. Introduction .....................................................................................................1 Long-Lasting Consequences of Systemic Inflammatory Event on Memory and Emotion ........................................................................................1 Neuroimmune Activation after Systemic Inflammatory Event and its Consequences on Behavior .........................................................................4 Memory: Types of Memory, Circuits, and Molecular Mechanisms ........................8 Immune Modulation of Memory and Cognitive Functions ..................................10 Mechanisms of memory dysfunction after systemic inflammatory event ............14 Goal Dissertation Project and Summary Hypotheses ............................................17 Chapter II. Long-lasting changes in memory after subchronic immune challenge 20 Abstract ..................................................................................................................20 Introduction ............................................................................................................21 Materials and methods ...........................................................................................23 Results ....................................................................................................................27 Discussion ..............................................................................................................36 Figures....................................................................................................................43 Chapter III. Implications of subchronic immune challenge on long-lasting depressive-like and anxiety-like behaviors ...................................................................53 Abstract ..................................................................................................................53 Introduction ............................................................................................................54 Materials and methods ...........................................................................................56 Results ....................................................................................................................60 Discussion ..............................................................................................................66 Figures....................................................................................................................74 Chapter IV. Enduring and sex-specific changes in hippocampal gene expression after subchronic immune challenge ...............................................................................84 Abstract ..................................................................................................................84 Introduction ............................................................................................................85 Materials and methods ...........................................................................................87 Results ....................................................................................................................91 Discussion ............................................................................................................104 Tables ...................................................................................................................113
Recommended publications
  • Genetic Analysis of Retinopathy in Type 1 Diabetes

    Genetic Analysis of Retinopathy in Type 1 Diabetes

    Genetic Analysis of Retinopathy in Type 1 Diabetes by Sayed Mohsen Hosseini A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Institute of Medical Science University of Toronto © Copyright by S. Mohsen Hosseini 2014 Genetic Analysis of Retinopathy in Type 1 Diabetes Sayed Mohsen Hosseini Doctor of Philosophy Institute of Medical Science University of Toronto 2014 Abstract Diabetic retinopathy (DR) is a leading cause of blindness worldwide. Several lines of evidence suggest a genetic contribution to the risk of DR; however, no genetic variant has shown convincing association with DR in genome-wide association studies (GWAS). To identify common polymorphisms associated with DR, meta-GWAS were performed in three type 1 diabetes cohorts of White subjects: Diabetes Complications and Control Trial (DCCT, n=1304), Wisconsin Epidemiologic Study of Diabetic Retinopathy (WESDR, n=603) and Renin-Angiotensin System Study (RASS, n=239). Severe (SDR) and mild (MDR) retinopathy outcomes were defined based on repeated fundus photographs in each study graded for retinopathy severity on the Early Treatment Diabetic Retinopathy Study (ETDRS) scale. Multivariable models accounted for glycemia (measured by A1C), diabetes duration and other relevant covariates in the association analyses of additive genotypes with SDR and MDR. Fixed-effects meta- analysis was used to combine the results of GWAS performed separately in WESDR, ii RASS and subgroups of DCCT, defined by cohort and treatment group. Top association signals were prioritized for replication, based on previous supporting knowledge from the literature, followed by replication in three independent white T1D studies: Genesis-GeneDiab (n=502), Steno (n=936) and FinnDiane (n=2194).
  • Comparative Transcriptome Analyses Indicate Molecular Homology of Zebrafish Swimbladder and Mammalian Lung

    Comparative Transcriptome Analyses Indicate Molecular Homology of Zebrafish Swimbladder and Mammalian Lung

    Comparative Transcriptome Analyses Indicate Molecular Homology of Zebrafish Swimbladder and Mammalian Lung Weiling Zheng1, Zhengyuan Wang1, John E. Collins2, Robert M. Andrews2, Derek Stemple2, Zhiyuan Gong1* 1 Department of Biological Sciences, National University of Singapore, Singapore, Singapore, 2 Vertebrate Development and Genetics, Wellcome Trust Genome Campus, Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom Abstract The fish swimbladder is a unique organ in vertebrate evolution and it functions for regulating buoyancy in most teleost species. It has long been postulated as a homolog of the tetrapod lung, but the molecular evidence is scarce. In order to understand the molecular function of swimbladder as well as its relationship with lungs in tetrapods, transcriptomic analyses of zebrafish swimbladder were carried out by RNA-seq. Gene ontology classification showed that genes in cytoskeleton and endoplasmic reticulum were enriched in the swimbladder. Further analyses depicted gene sets and pathways closely related to cytoskeleton constitution and regulation, cell adhesion, and extracellular matrix. Several prominent transcription factor genes in the swimbladder including hoxc4a, hoxc6a, hoxc8a and foxf1 were identified and their expressions in developing swimbladder during embryogenesis were confirmed. By comparison of enriched transcripts in the swimbladder with those in human and mouse lungs, we established the resemblance of transcriptome of the zebrafish swimbladder and mammalian lungs. Based on the transcriptomic data of zebrafish swimbladder, the predominant functions of swimbladder are in its epithelial and muscular tissues. Our comparative analyses also provide molecular evidence of the relatedness of the fish swimbladder and mammalian lung. Citation: Zheng W, Wang Z, Collins JE, Andrews RM, Stemple D, et al.
  • Network Medicine Approach for Analysis of Alzheimer's Disease Gene Expression Data

    Network Medicine Approach for Analysis of Alzheimer's Disease Gene Expression Data

    International Journal of Molecular Sciences Article Network Medicine Approach for Analysis of Alzheimer’s Disease Gene Expression Data David Cohen y, Alexander Pilozzi y and Xudong Huang * Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA; [email protected] (D.C.); [email protected] (A.P.) * Correspondence: [email protected]; Tel./Fax: +1-617-724-9778 These authors contributed equally to this work. y Received: 15 November 2019; Accepted: 30 December 2019; Published: 3 January 2020 Abstract: Alzheimer’s disease (AD) is the most widespread diagnosed cause of dementia in the elderly. It is a progressive neurodegenerative disease that causes memory loss as well as other detrimental symptoms that are ultimately fatal. Due to the urgent nature of this disease, and the current lack of success in treatment and prevention, it is vital that different methods and approaches are applied to its study in order to better understand its underlying mechanisms. To this end, we have conducted network-based gene co-expression analysis on data from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database. By processing and filtering gene expression data taken from the blood samples of subjects with varying disease states and constructing networks based on that data to evaluate gene relationships, we have been able to learn about gene expression correlated with the disease, and we have identified several areas of potential research interest. Keywords: Alzheimer’s disease; network medicine; gene expression; neurodegeneration; neuroinflammation 1. Introduction Alzheimer’s disease (AD) is the most widespread diagnosed cause of dementia in the elderly [1].
  • Downloaded the Normalized Gene Expression Profile from the GEO (Package Geoquery (5))

    Downloaded the Normalized Gene Expression Profile from the GEO (Package Geoquery (5))

    1 SUPPLEMENTARY MATERIALS 2 Supplementary Methods 3 Study design 4 The workflow to identify and validate the TME risk score and TME subtypes in gastric cancer is 5 depicted in Fig. S1. We first estimated the absolute abundance levels of the major stromal and 6 immune cell types in the TME using bulk gene expression data, and assessed the prognostic 7 effect of these cells in a discovery cohort. Next, we constructed a TME risk score and validated it 8 in two independent gene expression validation cohorts and three immunohistochemistry 9 validation cohorts. Finally, we stratified patients into four TME subtypes and examined their 10 genomic and molecular features and relation to established molecular subtypes. 11 Gene expression data 12 To explore the prognostic landscape of the TME, we used four gene expression profile (GEP) 13 datasets of resected gastric cancer patients with publicly available clinical information, namely, 14 ACRG (GSE62254) (1), GSE15459 (2), GSE84437 (3), and TCGA stomach adenocarcinoma 15 (STAD). Specifically, the raw microarray data in the ACRG cohort and GSE15459 were retrieved 16 from the Gene Expression Omnibus (GEO), and normalized by the RMA algorithm (package affy) 17 using custom chip definition files (Brainarray version 23 (4)) that convert Affymetrix probesets to 18 Entrez gene IDs. For GSE84437 dataset, which was measured by the Illumina platform, we 19 downloaded the normalized gene expression profile from the GEO (package GEOquery (5)). The 20 Illumina probes were also mapped to Entrez genes. For multiple probes mapping to the same 21 Entrez gene, we selected the one with the maximum mean expression level as the surrogate for 22 the Entrez gene using the function of collapseRows (6) (package WGCNA).
  • Supplemental Table S1

    Supplemental Table S1

    Entrez Gene Symbol Gene Name Affymetrix EST Glomchip SAGE Stanford Literature HPA confirmed Gene ID Profiling profiling Profiling Profiling array profiling confirmed 1 2 A2M alpha-2-macroglobulin 0 0 0 1 0 2 10347 ABCA7 ATP-binding cassette, sub-family A (ABC1), member 7 1 0 0 0 0 3 10350 ABCA9 ATP-binding cassette, sub-family A (ABC1), member 9 1 0 0 0 0 4 10057 ABCC5 ATP-binding cassette, sub-family C (CFTR/MRP), member 5 1 0 0 0 0 5 10060 ABCC9 ATP-binding cassette, sub-family C (CFTR/MRP), member 9 1 0 0 0 0 6 79575 ABHD8 abhydrolase domain containing 8 1 0 0 0 0 7 51225 ABI3 ABI gene family, member 3 1 0 1 0 0 8 29 ABR active BCR-related gene 1 0 0 0 0 9 25841 ABTB2 ankyrin repeat and BTB (POZ) domain containing 2 1 0 1 0 0 10 30 ACAA1 acetyl-Coenzyme A acyltransferase 1 (peroxisomal 3-oxoacyl-Coenzyme A thiol 0 1 0 0 0 11 43 ACHE acetylcholinesterase (Yt blood group) 1 0 0 0 0 12 58 ACTA1 actin, alpha 1, skeletal muscle 0 1 0 0 0 13 60 ACTB actin, beta 01000 1 14 71 ACTG1 actin, gamma 1 0 1 0 0 0 15 81 ACTN4 actinin, alpha 4 0 0 1 1 1 10700177 16 10096 ACTR3 ARP3 actin-related protein 3 homolog (yeast) 0 1 0 0 0 17 94 ACVRL1 activin A receptor type II-like 1 1 0 1 0 0 18 8038 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 1 0 0 0 0 19 8751 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 1 0 0 0 0 20 8728 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 1 0 0 0 0 21 81792 ADAMTS12 ADAM metallopeptidase with thrombospondin type 1 motif, 12 1 0 0 0 0 22 9507 ADAMTS4 ADAM metallopeptidase with thrombospondin type 1
  • New PDF Document

    New PDF Document

    888.267.4436 [email protected] www.origene.com Name:CLDN6 mouse monoclonal antibody, clone OTI3E3 (formerly 3E3) Catalog: TA507011 Product Data Sheet - TRUEMAB Components: • CLDN6 mouse monoclonal antibody, clone OTI3E3 (formerly 3E3) (TA507011) • WB positive control also available: 20ug myc-DDK tagged CLDN6 HEK293T over-expression lysate lyophilized in RIPA buffer (LC412034). (Reconstitute into 20ul of 1x SDS sample buffer before loading; load 5ul per lane as WB control or as desired) Amount: 100ul Immunogen: Full length human recombinant protein of human CLDN6(NP_067018) produced in HEK293T cell. Host: Mouse Isotype: IgG1 Species Reactivity: Human Guaranteed WB, IF Applications: Suggested WB 1:1000, IF 1:100, Dilutions: Concentration: 1 mg/ml Buffer: PBS (PH 7.3) containing 1% BSA, 50% glycerol and 0.02% sodium azide. Purification: Purified from mouse ascites fluids by affinity chromatography Storage Condition: Shipped at -20C or with ice packs. Upon delivery store at -20C. Dilute in PBS (pH7.3) if necessary. Stable for 12 months from date of receipt. Avoid repeated freeze-thaws. Target Target Name: Homo sapiens claudin 6 (CLDN6) Alternative Name: OTTHUMP00000159248; claudin 6 Database Link: NP_067018 Entrez Gene 9074 Human Function: Tight junctions represent one mode of cell-to-cell adhesion in epithelial or endothelial cell sheets, forming continuous seals around cells and serving as a physical barrier to prevent solutes and water from passing freely through the paracellular space. These junctions are comprised of sets of continuous networking strands in the outwardly facing cytoplasmic leaflet, with complementary grooves in the inwardly facing extracytoplasmic leaflet. This gene encodes a component of tight junction strands, which This product is to be used for laboratory only.
  • Steroid-Dependent Regulation of the Oviduct: a Cross-Species Transcriptomal Analysis

    Steroid-Dependent Regulation of the Oviduct: a Cross-Species Transcriptomal Analysis

    University of Kentucky UKnowledge Theses and Dissertations--Animal and Food Sciences Animal and Food Sciences 2015 Steroid-dependent regulation of the oviduct: A cross-species transcriptomal analysis Katheryn L. Cerny University of Kentucky, [email protected] Right click to open a feedback form in a new tab to let us know how this document benefits ou.y Recommended Citation Cerny, Katheryn L., "Steroid-dependent regulation of the oviduct: A cross-species transcriptomal analysis" (2015). Theses and Dissertations--Animal and Food Sciences. 49. https://uknowledge.uky.edu/animalsci_etds/49 This Doctoral Dissertation is brought to you for free and open access by the Animal and Food Sciences at UKnowledge. It has been accepted for inclusion in Theses and Dissertations--Animal and Food Sciences by an authorized administrator of UKnowledge. For more information, please contact [email protected]. STUDENT AGREEMENT: I represent that my thesis or dissertation and abstract are my original work. Proper attribution has been given to all outside sources. I understand that I am solely responsible for obtaining any needed copyright permissions. I have obtained needed written permission statement(s) from the owner(s) of each third-party copyrighted matter to be included in my work, allowing electronic distribution (if such use is not permitted by the fair use doctrine) which will be submitted to UKnowledge as Additional File. I hereby grant to The University of Kentucky and its agents the irrevocable, non-exclusive, and royalty-free license to archive and make accessible my work in whole or in part in all forms of media, now or hereafter known.
  • Emerging Roles for Multifunctional Ion Channel Auxiliary Subunits in Cancer T ⁎ Alexander S

    Emerging Roles for Multifunctional Ion Channel Auxiliary Subunits in Cancer T ⁎ Alexander S

    Cell Calcium 80 (2019) 125–140 Contents lists available at ScienceDirect Cell Calcium journal homepage: www.elsevier.com/locate/ceca Emerging roles for multifunctional ion channel auxiliary subunits in cancer T ⁎ Alexander S. Hawortha,b, William J. Brackenburya,b, a Department of Biology, University of York, Heslington, York, YO10 5DD, UK b York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK ARTICLE INFO ABSTRACT Keywords: Several superfamilies of plasma membrane channels which regulate transmembrane ion flux have also been Auxiliary subunit shown to regulate a multitude of cellular processes, including proliferation and migration. Ion channels are Cancer typically multimeric complexes consisting of conducting subunits and auxiliary, non-conducting subunits. Calcium channel Auxiliary subunits modulate the function of conducting subunits and have putative non-conducting roles, further Chloride channel expanding the repertoire of cellular processes governed by ion channel complexes to processes such as trans- Potassium channel cellular adhesion and gene transcription. Given this expansive influence of ion channels on cellular behaviour it Sodium channel is perhaps no surprise that aberrant ion channel expression is a common occurrence in cancer. This review will − focus on the conducting and non-conducting roles of the auxiliary subunits of various Ca2+,K+,Na+ and Cl channels and the burgeoning evidence linking such auxiliary subunits to cancer. Several subunits are upregu- lated (e.g. Cavβ,Cavγ) and downregulated (e.g. Kvβ) in cancer, while other subunits have been functionally implicated as oncogenes (e.g. Navβ1,Cavα2δ1) and tumour suppressor genes (e.g. CLCA2, KCNE2, BKγ1) based on in vivo studies. The strengthening link between ion channel auxiliary subunits and cancer has exposed these subunits as potential biomarkers and therapeutic targets.
  • Supplementary Table 1: Adhesion Genes Data Set

    Supplementary Table 1: Adhesion Genes Data Set

    Supplementary Table 1: Adhesion genes data set PROBE Entrez Gene ID Celera Gene ID Gene_Symbol Gene_Name 160832 1 hCG201364.3 A1BG alpha-1-B glycoprotein 223658 1 hCG201364.3 A1BG alpha-1-B glycoprotein 212988 102 hCG40040.3 ADAM10 ADAM metallopeptidase domain 10 133411 4185 hCG28232.2 ADAM11 ADAM metallopeptidase domain 11 110695 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 195222 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 165344 8751 hCG20021.3 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 189065 6868 null ADAM17 ADAM metallopeptidase domain 17 (tumor necrosis factor, alpha, converting enzyme) 108119 8728 hCG15398.4 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 117763 8748 hCG20675.3 ADAM20 ADAM metallopeptidase domain 20 126448 8747 hCG1785634.2 ADAM21 ADAM metallopeptidase domain 21 208981 8747 hCG1785634.2|hCG2042897 ADAM21 ADAM metallopeptidase domain 21 180903 53616 hCG17212.4 ADAM22 ADAM metallopeptidase domain 22 177272 8745 hCG1811623.1 ADAM23 ADAM metallopeptidase domain 23 102384 10863 hCG1818505.1 ADAM28 ADAM metallopeptidase domain 28 119968 11086 hCG1786734.2 ADAM29 ADAM metallopeptidase domain 29 205542 11085 hCG1997196.1 ADAM30 ADAM metallopeptidase domain 30 148417 80332 hCG39255.4 ADAM33 ADAM metallopeptidase domain 33 140492 8756 hCG1789002.2 ADAM7 ADAM metallopeptidase domain 7 122603 101 hCG1816947.1 ADAM8 ADAM metallopeptidase domain 8 183965 8754 hCG1996391 ADAM9 ADAM metallopeptidase domain 9 (meltrin gamma) 129974 27299 hCG15447.3 ADAMDEC1 ADAM-like,
  • The Role of the X Chromosome in Embryonic and Postnatal Growth

    The Role of the X Chromosome in Embryonic and Postnatal Growth

    The role of the X chromosome in embryonic and postnatal growth Daniel Mark Snell A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy of University College London. Francis Crick Institute/Medical Research Council National Institute for Medical Research University College London January 28, 2018 2 I, Daniel Mark Snell, 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 work. Abstract Women born with only a single X chromosome (XO) have Turner syndrome (TS); and they are invariably of short stature. XO female mice are also small: during embryogenesis, female mice with a paternally-inherited X chromosome (XPO) are smaller than XX littermates; whereas during early postnatal life, both XPO and XMO (maternal) mice are smaller than their XX siblings. Here I look to further understand the genetic bases of these phenotypes, and potentially inform areas of future investigation into TS. Mouse pre-implantation embryos preferentially silence the XP via the non-coding RNA Xist.XPO embryos are smaller than XX littermates at embryonic day (E) 10.5, whereas XMO embryos are not. Two possible hypotheses explain this obser- vation. Inappropriate expression of Xist in XPO embryos may cause transcriptional silencing of the single X chromosome and result in embryos nullizygous for X gene products. Alternatively, there could be imprinted genes on the X chromosome that impact on growth and manifest in growth retarded XPO embryos. In contrast, dur- ing the first three weeks of postnatal development, both XPO and XMO mice show a growth deficit when compared with XX littermates.
  • Association of Gene Ontology Categories with Decay Rate for Hepg2 Experiments These Tables Show Details for All Gene Ontology Categories

    Association of Gene Ontology Categories with Decay Rate for Hepg2 Experiments These Tables Show Details for All Gene Ontology Categories

    Supplementary Table 1: Association of Gene Ontology Categories with Decay Rate for HepG2 Experiments These tables show details for all Gene Ontology categories. Inferences for manual classification scheme shown at the bottom. Those categories used in Figure 1A are highlighted in bold. Standard Deviations are shown in parentheses. P-values less than 1E-20 are indicated with a "0". Rate r (hour^-1) Half-life < 2hr. Decay % GO Number Category Name Probe Sets Group Non-Group Distribution p-value In-Group Non-Group Representation p-value GO:0006350 transcription 1523 0.221 (0.009) 0.127 (0.002) FASTER 0 13.1 (0.4) 4.5 (0.1) OVER 0 GO:0006351 transcription, DNA-dependent 1498 0.220 (0.009) 0.127 (0.002) FASTER 0 13.0 (0.4) 4.5 (0.1) OVER 0 GO:0006355 regulation of transcription, DNA-dependent 1163 0.230 (0.011) 0.128 (0.002) FASTER 5.00E-21 14.2 (0.5) 4.6 (0.1) OVER 0 GO:0006366 transcription from Pol II promoter 845 0.225 (0.012) 0.130 (0.002) FASTER 1.88E-14 13.0 (0.5) 4.8 (0.1) OVER 0 GO:0006139 nucleobase, nucleoside, nucleotide and nucleic acid metabolism3004 0.173 (0.006) 0.127 (0.002) FASTER 1.28E-12 8.4 (0.2) 4.5 (0.1) OVER 0 GO:0006357 regulation of transcription from Pol II promoter 487 0.231 (0.016) 0.132 (0.002) FASTER 6.05E-10 13.5 (0.6) 4.9 (0.1) OVER 0 GO:0008283 cell proliferation 625 0.189 (0.014) 0.132 (0.002) FASTER 1.95E-05 10.1 (0.6) 5.0 (0.1) OVER 1.50E-20 GO:0006513 monoubiquitination 36 0.305 (0.049) 0.134 (0.002) FASTER 2.69E-04 25.4 (4.4) 5.1 (0.1) OVER 2.04E-06 GO:0007050 cell cycle arrest 57 0.311 (0.054) 0.133 (0.002)
  • Mouse Steap4 Conditional Knockout Project (CRISPR/Cas9)

    Mouse Steap4 Conditional Knockout Project (CRISPR/Cas9)

    https://www.alphaknockout.com Mouse Steap4 Conditional Knockout Project (CRISPR/Cas9) Objective: To create a Steap4 conditional knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Steap4 gene (NCBI Reference Sequence: NM_054098 ; Ensembl: ENSMUSG00000012428 ) is located on Mouse chromosome 5. 5 exons are identified, with the ATG start codon in exon 2 and the TAA stop codon in exon 5 (Transcript: ENSMUST00000115421). Exon 2~3 will be selected as conditional knockout region (cKO region). Deletion of this region should result in the loss of function of the Mouse Steap4 gene. To engineer the targeting vector, homologous arms and cKO region will be generated by PCR using BAC clone RP23-212F6 as template. Cas9, gRNA and targeting vector will be co-injected into fertilized eggs for cKO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Mice homozygous for a knock-out allele exhibit adipose accumulation, oxidative stress, increased liver weight, lower metabolic rate, hypoactivity, insulin resistance, glucose intolerance, mild hyperglycemia and dyslipidemia. Exon 2 starts from about 100% of the coding region. The knockout of Exon 2~3 will result in frameshift of the gene. The size of intron 1 for 5'-loxP site insertion: 14897 bp, and the size of intron 3 for 3'-loxP site insertion: 1385 bp. The size of effective cKO region: ~2082 bp. The cKO region does not have any other known gene. Page 1 of 7 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele 5' gRNA region gRNA region 3' 1 2 3 4 5 Targeting vector Targeted allele Constitutive KO allele (After Cre recombination) Legends Exon of mouse Steap4 Homology arm cKO region loxP site Page 2 of 7 https://www.alphaknockout.com Overview of the Dot Plot Window size: 10 bp Forward Reverse Complement Sequence 12 Note: The sequence of homologous arms and cKO region is aligned with itself to determine if there are tandem repeats.