Signature Redacted Harvard-MIT Program in Health Sciences and 'Kchnology September 29, 2015

Total Page:16

File Type:pdf, Size:1020Kb

Signature Redacted Harvard-MIT Program in Health Sciences and 'Kchnology September 29, 2015 A single-cell perspective on infection by Nathan Scott Haseley B.S. Rochester Institute of Technology (2009) Submitted to the Harvard-MIT Program In Health Sciences and Technology in partial fulfillnent of the requirements for the degree of Doctor of Philosophy in Bioinformatics at the MASSACHUSETTS INSTITUTE OF TECHNOLOGY Februrary 2016 Massachusetts Institute of Technology 2016. All rights reserved. Author .................................... Signature redacted Harvard-MIT Program In Health Sciences and 'kchnology September 29, 2015 Signature redacted C ertified by .......... ................... ............. \ Deborah T. Hung MD, PhD, Associate Professor Thesis Supervisor Signature redacted A ccepted by ........ .. .. .. .. ... .. ... .. ... .. .. ... .. .. Emery N. Brown MD, PhD/Director, Harvard T Program in Health Sciences and Technology/Professor of Computational Neuroscience and Health Sciences and Technology MASSACHUSES INSTITUTE OF TECHN0LOGY MAR 142016 LIBRARI S '2 A single-cell perspective on infection by Nathan Scott Haseley Submitted to the Harvard-MIT Program In Health Sciences and Technology on September 29, 2015, in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Bioinformatics Abstract The clinical course of infection is ultimately determined by a series of cellular interactions between invading pathogens and host immune cells. It has long been understood that these interactions, even when they occur in tissue culture models, give rise to a wide variety of different outcomes, some beneficial to the host, others to the pathogen. These cellular interactions, however, are typically studied at a bulk level; masking this cell-to-cell variation, losing important information about the full range of possible host-pathogen interactions, and leaving the mechanistic basis for these different outcomes largely unexplored. Here, we present a system that combines single-cell RNA sequencing with fluorescent markers of infection outcome to directly correlate host transcription signatures with infection outcome at the single cell level. Applying this system to the well-characterized model of Salmonella enterica infection of mouse macrophages, we found: 1) Unique transcription signatures associated with bacterial exposure and bacterial infection, 2) Sustained high levels of heterogene- ity in immune pathways in infected macrophages, and 3) A novel subpopulation of macrophages characterized by high expression of the Type I Interferon response after infection. Upon further investigation we found that this heterogeneity in the host Type I Interferon response was the result of heterogeneity in the population of infecting bacteria, namely in the extent of PhoPQ-mediated LPS modifications. This work highlights the importance of heterogeneity as a characteristic of bac- terial populations that can influence the host immune response. It also demonstrates the benefits of examining infection with single-cell resolution. Thesis Supervisor: Deborah T. Hung Title: MD, PhD, Associate Professor 3 4 Acknowledgements This thesis is a product of all of the support, care, and instruction that I have received over many years. There are many people to thank that have been instrumental in my graduate career and in getting me to this point. I am sure that I did not mention everyone here but nevertheless, I would like to say thank you. First, I would like to thank my wife, Psalm, for all her support and encouragement throughout graduate school and for making Boston into a home for both of us. I would like to thank my parents for their support and wisdom throughout the years. Also the numerous friends that I've had in Boston. I owe a debt to the entire community at CityLife Presbyterian Church for their support and prayers, and particularly to my community group. Thanks as well to Sam, Whitney, Trevor, and Megan for numerous game nights and other times of hanging out that were essential for sanity. I would like to thank my adviser Deb, for teaching me how to think like a scientist and all current and past members of the Hung lab for helpful discussions, encouragement when things weren't working, and for making the Broad Institute a great place to work. I would also like to thank all of our collaborators in the Regev and Xavier labs. I would additionally like to thank my previous mentor Dr. Ferran, for getting me started in science and old members of the Ferran lab for their still-constant friendship. Finally, I would like to thank all the members of my committee, all of the HST staff for their help through this process, and my classmates, particularly all of my fellow HST BIG students for feedback, friendship, and keeping me interested in science outside my own project. 5 6 Contents 1 Introduction 15 1.1 O verview ....... ............... ............... ..... 15 1.2 S. enterica pathophysiology .. .......... .......... ......... 17 1.3 Extracellular host-pathogen interactions ... ....... ........ ....... 18 1.3.1 The role of diverse inicroenvironments in promoting heterogeneity .... .. 18 1.3.2 Heterogeneity as a mechanism to control the host inflammatory response . 20 1.4 Intracellular host-pathogen interactions: A battle between two highly adaptable cells 21 1.5 Heterogeneity in macrophage and bacterial populations ........ ......... 25 1.6 The need for a new approach to study infection ......... ......... ... 26 2 Examining S. enterica-Macrophage Interactions at the Single-Cell Level 29 2.1 A ttributions .... ........ ....... ....... ........ ....... 29 2.2 Introduction ... ....... ....... ........ ....... ........ 29 2.3 Results . ....... .... .......... ......... ....... 30 2.3.1 Heterogeneous outcomes of S. enterica-macrophage encounters ........ 30 2.3.2 Single-cell RNA-Seq accurately captures host transcriptional states after bac- terial exposure . ........ ......... ........ ......... 31 2.3.3 Single-cell RNA-Seq identifies transcriptional changes associated with extra- cellular and intracellular bacterial detection .......... ......... 35 2.3.4 Bimodal induction of Type I IFN response genes in infected macrophages .. 37 2.3.5 Infected macrophages display high cell-to-cell variation in genes from immune response pathways .. ......... ........ ......... ..... 40 2.4 D iscussion .......... ............... ............... .. 43 7 2.4.1 A general approach to characterize the transcriptional underpinnings of phe- notypic heterogeneity in host-pathogen encounters ........ ..... 43 2.4.2 . A single-cell resolution map of S. typhimuriumr-inacrophageinteractions . 43 2.5 Materials and Methods ....... .... ... ... ... ... 4 4 2.5.1 Mice, cell lines and bacterial strains . ..... .... ... .. ... ... 44 2.5.2 Cell and bacterial cultures, single-cell sorting, and analysis . ... ... ... 45 2.5.3 Imaging assay protocol .. ...... ....... ...... .. .. .. .. 45 2.5.4 Image acquisition and analysis .... ....... ..... ... .. .. 45 2.5.5 Single-cell expression profiling ....... ...... .... .. .. .. .. 46 2.5.6 cDNA synthesis, amplification and library construction . .. .. .. .. .. 46 2.5.7 Transcript quantification . ...... ....... ...... ... ... .. 47 2.5.8 Quality filters and statistics . ..... ..... .... ... .. ... ... 47 2.5.9 Comparison of methods for differential expression analysis . .. .. .. .. 48 2.5.10 Differential expression analysis ...... ...... .... ... ... ... 48 2.5.11 P C A ... ...... ....... ...... ...... ... .. .. .. .. 49 2.5.12 Identifying genes responding to extracellular or intracellular ba(teria (Clusters I and II) ..... ...... ...... .. ..... 49 2.5.13 Identification of co-regulated gene clusters (Clusters III, IV, and V) . .... 50 2.5.14 Correlation plots ... ..... .... ..... .... .... ..... ... 51 2.5.15 H eatm aps .. ..... ...... ..... ..... ...... ..... ... 51 2.5.16 Single-molecule RNA-flow FISH ......... ......... ....... 51 2.5.17 Estimating gene variance scores . ..... ..... ...... ..... ... 51 2.5.18 Identification of low and high variance pathways in infected inacrophages . 52 2.5.19 Comparison of variance scores between exposure- and infection-induced gene ...... .............. ........ sets ..... ..... .... 52 2.5.20 Cell expression density plQts . ..... ..... ...... ..... ..... 52 2.5.21 D atasets ... ..... ...... ..... ..... ...... ..... ... 53 3 The Mechanisms Behind Heterogeneity in the Type I IFN Response 55 3.1 A ttributions .. ..... .... .... ..... .... ..... .... ..... .. 55 3.2 Introduction .. ...... ....... ....... ...... ....... ..... 55 3.3 R esu lts .. ......... ......... ......... ......... ..... 56 8 3.3.1 Intracellular TLR4 signaling through TRIF and IRF3 determines the expres- sion of the Type I IFN response in infected macrophages ...... ...... 56 3.3.2 Live bacteria, but not LPS-coated beads, elicit a variable Type I IFN response in infected macrophages ............. ............. .... 58 3.3.3 Variation in the host Type I IFN response is driven by bimodal activity of the bacterial PhoPQ two-component system in infecting bacteria ....... 59 3.3.4 Intracellular recognition of PhoPQ-mediated LPS modifications results in in- duction of the Type I IFN response ........................ 62 3.3.5 PhoPQ-mediated LPS modifications impact the in vivo Type I IFN response and infection outcome ............................... 64 3.4 D iscussion ...... ............................. ....... 67 3.4.1 Heterogeneity of pathogen populations as a mechanism to shape the host im mune response .................................. 67 3.4.2 Studies of the immune response in the context of heterogeneous
Recommended publications
  • Tristetraprolin Limits Inflammatory Cytokine Production in Tumor-Associated Macrophages in an Mrna Decay− Independent Manner
    Published OnlineFirst July 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0205 Cancer Microenvironment and Immunology Research Tristetraprolin Limits Inflammatory Cytokine Production in Tumor-Associated Macrophages in an mRNA Decay–Independent Manner Franz Kratochvill1,2, Nina Gratz1, Joseph E. Qualls1,2, Lee-Ann Van De Velde1,2, Hongbo Chi2, Pavel Kovarik3, and Peter J. Murray1,2 Abstract Tristetraprolin (TTP) is an inducible zinc finger AU-rich (TAM). However, TTP's effects on AU-rich mRNA stability RNA-binding protein essential for enforcing degradation of were negligible and limited by constitutive p38a MAPK activ- mRNAs encoding inflammatory chemokines and cytokines. ity, which was the main driver of proinflammatory cytokine Most studies on TTP center on the connection between mRNA production in TAMs at the posttranscriptional level. Instead, half-life and inflammatory output, because loss of TTP ampli- elimination of TTP caused excessive protein production of fies inflammation by increasing the stability of AU-rich inflammatory mediators, suggesting TTP-dependent transla- mRNAs. Here, we focused on how TTP controls cytokine and tional suppression of AU-rich mRNAs. Manipulation of the chemokine production in the nonresolving inflammation of p38a–TTP axis in macrophages has significant effects on the cancer using tissue-specific approaches. In contrast with mod- growth of tumors and therefore represents a means to mani- el in vitro macrophage systems, we found constitutive TTP pulate inflammation in the tumor microenvironment. Cancer expression in late-stage tumor-associated macrophages Res; 75(15); 1–11. Ó2015 AACR. Introduction TLR signaling phosphorylates TTP thereby blocking its function and sustaining TNF output (9, 10).
    [Show full text]
  • Dynamic Regulation of FGF23 by Fam20c Phosphorylation, Galnac-T3 Glycosylation, and Furin Proteolysis
    Dynamic regulation of FGF23 by Fam20C phosphorylation, GalNAc-T3 glycosylation, and furin proteolysis Vincent S. Tagliabraccia, James L. Engela,1, Sandra E. Wileya, Junyu Xiaoa, David J. Gonzaleza,b, Hitesh Nidumanda Appaiahc, Antonius Kollerd, Victor Nizetb,e, Kenneth E. Whitec, and Jack E. Dixona,f,g,2 Departments of aPharmacology, bPediatrics, fCellular and Molecular Medicine, and gChemistry and Biochemistry and eSkaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093; cDepartment of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202; and dStony Brook University Proteomics Center, School of Medicine, Stony Brook University, Stony Brook, NY 11794 Contributed by Jack E. Dixon, February 8, 2014 (sent for review January 23, 2014) The family with sequence similarity 20, member C (Fam20C) has life. Nonlethal cases have also been reported, and these patients recently been identified as the Golgi casein kinase. Fam20C phosphor- develop hypophosphatemia as a result of elevated levels of the ylates secreted proteins on Ser-x-Glu/pSer motifs and loss-of-function phosphate-regulating hormone fibroblast growth factor 23 (FGF23) mutations in the kinase cause Raine syndrome, an often-fatal osteo- (12). Additionally, Fam20C knockout (KO) mice develop renal sclerotic bone dysplasia. Fam20C is potentially an upstream regulator phosphate wasting due to an increase in circulating bioactive of the phosphate-regulating hormone fibroblast growth factor 23 FGF23 as well as severe hypophosphatemic rickets (13, 14). (FGF23), because humans with FAM20C mutations and Fam20C KO Thus, Fam20C has been proposed to be a regulator of FGF23; mice develop hypophosphatemia due to an increase in full-length, bi- however, the molecular mechanisms underlying the control of ologically active FGF23.
    [Show full text]
  • Datasheet PB1029 Anti-AEBP2 Antibody
    Product datasheet Anti-AEBP2 Antibody Catalog Number: PB1029 BOSTER BIOLOGICAL TECHNOLOGY Special NO.1, International Enterprise Center, 2nd Guanshan Road, Wuhan, China Web: www.boster.com.cn Phone: +86 27 67845390 Fax: +86 27 67845390 Email: [email protected] Basic Information Product Name Anti-AEBP2 Antibody Gene Name AEBP2 Source Rabbit IgG Species Reactivity human,mouse,rat Tested Application WB,IHC-P,ICC/IF,FCM Contents 500ug/ml antibody with PBS ,0.02% NaN3 , 1mg BSA and 50% glycerol. Immunogen E.coli-derived human AEBP2 recombinant protein (Position: K424-Q517). Human AEBP2 shares 98.8% amino acid (aa) sequence identity with mouse AEBP2. Purification Immunogen affinity purified. Observed MW 54KD Dilution Ratios Western blot: 1:500-2000 Immunohistochemistry(Paraffin-embedded Section): 1:50-400 Immunocytochemistry/Immunofluorescence (ICC/IF): 1:50-400 Flow cytometry (FCM): 1-3μg/1x106 cells Storage 12 months from date of receipt,-20℃ as supplied.6 months 2 to 8℃ after reconstitution. Avoid repeated freezing and thawing Background Information Adipocyte Enhancer-Binding Protein is a zinc finger protein that in humans is encoded by the evolutionarily well-conserved gene AEBP2. This gene is mapped to 12p12.3. AEBP2 is a DNA-binding transcriptional repressor. It may regulate the migration and development of the neural crest cells through the PRC2-mediated epigenetic mechanism and is most likely a targeting protein for the mammalian PRC2 complex. Reference Anti-AEBP2 Antibody被引用在0文献中。 暂无引用 FOR RESEARCH USE ONLY. NOT FOR DIAGNOSTIC AND CLINICAL USE. 1 Product datasheet Anti-AEBP2 Antibody Catalog Number: PB1029 BOSTER BIOLOGICAL TECHNOLOGY Special NO.1, International Enterprise Center, 2nd Guanshan Road, Wuhan, China Web: www.boster.com.cn Phone: +86 27 67845390 Fax: +86 27 67845390 Email: [email protected] Selected Validation Data Figure 1.
    [Show full text]
  • Table 2. Significant
    Table 2. Significant (Q < 0.05 and |d | > 0.5) transcripts from the meta-analysis Gene Chr Mb Gene Name Affy ProbeSet cDNA_IDs d HAP/LAP d HAP/LAP d d IS Average d Ztest P values Q-value Symbol ID (study #5) 1 2 STS B2m 2 122 beta-2 microglobulin 1452428_a_at AI848245 1.75334941 4 3.2 4 3.2316485 1.07398E-09 5.69E-08 Man2b1 8 84.4 mannosidase 2, alpha B1 1416340_a_at H4049B01 3.75722111 3.87309653 2.1 1.6 2.84852656 5.32443E-07 1.58E-05 1110032A03Rik 9 50.9 RIKEN cDNA 1110032A03 gene 1417211_a_at H4035E05 4 1.66015788 4 1.7 2.82772795 2.94266E-05 0.000527 NA 9 48.5 --- 1456111_at 3.43701477 1.85785922 4 2 2.8237185 9.97969E-08 3.48E-06 Scn4b 9 45.3 Sodium channel, type IV, beta 1434008_at AI844796 3.79536664 1.63774235 3.3 2.3 2.75319499 1.48057E-08 6.21E-07 polypeptide Gadd45gip1 8 84.1 RIKEN cDNA 2310040G17 gene 1417619_at 4 3.38875643 1.4 2 2.69163229 8.84279E-06 0.0001904 BC056474 15 12.1 Mus musculus cDNA clone 1424117_at H3030A06 3.95752801 2.42838452 1.9 2.2 2.62132809 1.3344E-08 5.66E-07 MGC:67360 IMAGE:6823629, complete cds NA 4 153 guanine nucleotide binding protein, 1454696_at -3.46081884 -4 -1.3 -1.6 -2.6026947 8.58458E-05 0.0012617 beta 1 Gnb1 4 153 guanine nucleotide binding protein, 1417432_a_at H3094D02 -3.13334396 -4 -1.6 -1.7 -2.5946297 1.04542E-05 0.0002202 beta 1 Gadd45gip1 8 84.1 RAD23a homolog (S.
    [Show full text]
  • Pulmonary Fibrosis Associated with TINF2 Gene Mutation: Is Somatic Reversion Required?
    Pulmonary fibrosis associated with TINF2 gene mutation: is somatic reversion required? To the Editor: We read with great interest the case reported by FUKUHARA et al. [1] of a 43-year-old female patient with dyskeratosis congenita, pulmonary fibrosis and heterozygous mutation in TINF2 (telomerase repeat binding factor 1-interacting nuclear factor 2). TIN2, the TINF2 gene product, TERT (telomere reverse transcriptase) and TERC (telomerase RNA component) participate in the regulation of telomere elongation, in which mutations have been previously found to be associated with familial pulmonary fibrosis in adults [2]. Indeed mutations of SFTPC, coding for surfactant protein C, were initially described in children before being described in adults as old as 72 years of age who presented with familial pulmonary fibrosis [3]. However, we were surprised that a TINF2 mutation could be evidenced in an adult of that age. As highlighted by FUKUHARA et al. [1], patients with the TINF2 mutation present with severe haematological symptoms before 10 years of age [4]. As mentioned by FUKUHARA et al. [1], the identified mutation is probably not hypomorphic because it is a frame-shift deletion located in the mutational ‘‘hot spot’’ described previously. Furthermore, the patient presented with very short telomeres. The TINF2 mutation was probably inherited from her father because he had abnormal skin pigmentation and aplastic anaemia [1]. Re-analysis of the gene mutation sequencing could provide new hypotheses for this late disease onset. Indeed, the electrophoregram depicted in figure 1b in the study by FUKUHARA et al. [1] probably comes from a PCR product sub-cloned into an expression vector [5], and does not ensure that the deletion is at the heterozygous status usually seen in our patients (fig.
    [Show full text]
  • FAM158A Polyclonal Antibody Catalog Number:23919-1-AP
    For Research Use Only FAM158A Polyclonal antibody www.ptgcn.com Catalog Number:23919-1-AP Catalog Number: GenBank Accession Number: Recommended Dilutions: Basic Information 23919-1-AP BC002491 WB 1:200-1:1000 Size: GeneID (NCBI): IHC 1:20-1:200 500 μg/ml 51016 IF 1:20-1:200 Source: Full Name: Rabbit family with sequence similarity 158, Isotype: member A IgG Calculated MW: Purification Method: 208 aa, 23 kDa Antigen Affinity purified Observed MW: Immunogen Catalog Number: 23 kDa AG20744 Applications Tested Applications: Positive Controls: IF, IHC, WB,ELISA WB : HEK-293 cells; Species Specificity: IHC : human pancreas tissue; human Note-IHC: suggested antigen retrieval with IF : HEK-293 cells; TE buffer pH 9.0; (*) Alternatively, antigen retrieval may be performed with citrate buffer pH 6.0 UPF0172 protein FAM158A, also known as c14orf122 or CGI112, is a protein that in humans is encoded by the Background Information FAM158A gene located on chromosome 14q11.2. Human FAM158A and its paralogs in other species are part of the uncharacterized protein family UPF0172 family, which is a subset of the JAB1/Mov34/MPN/PAD-1 ubiquitin protease protein family. The MPN superfamily contributes to ubiquitination and de-ubiquitination activity within the cell. The UPF0172 subset no longer has a functional ubiquitination domain and the function is uncharacterized. Fam158a is nearly ubiquitously expressed throughout the human body. The homolog in mice also shows expression throughout the entire body. Several micro-arrays demonstrate the variable expression of Fam158a in response to other factors and in various cancer types. None of this information gives any indication of a specific function but the wide expression of the gene and its high conservation indicate that Fam158a plays an important role in cellular function.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Genetics of Familial Non-Medullary Thyroid Carcinoma (FNMTC)
    cancers Review Genetics of Familial Non-Medullary Thyroid Carcinoma (FNMTC) Chiara Diquigiovanni * and Elena Bonora Unit of Medical Genetics, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; [email protected] * Correspondence: [email protected]; Tel.: +39-051-208-8418 Simple Summary: Non-medullary thyroid carcinoma (NMTC) originates from thyroid follicular epithelial cells and is considered familial when occurs in two or more first-degree relatives of the patient, in the absence of predisposing environmental factors. Familial NMTC (FNMTC) cases show a high genetic heterogeneity, thus impairing the identification of pivotal molecular changes. In the past years, linkage-based approaches identified several susceptibility loci and variants associated with NMTC risk, however only few genes have been identified. The advent of next-generation sequencing technologies has improved the discovery of new predisposing genes. In this review we report the most significant genes where variants predispose to FNMTC, with the perspective that the integration of these new molecular findings in the clinical data of patients might allow an early detection and tailored therapy of the disease, optimizing patient management. Abstract: Non-medullary thyroid carcinoma (NMTC) is the most frequent endocrine tumor and originates from the follicular epithelial cells of the thyroid. Familial NMTC (FNMTC) has been defined in pedigrees where two or more first-degree relatives of the patient present the disease in absence of other predisposing environmental factors. Compared to sporadic cases, FNMTCs are often multifocal, recurring more frequently and showing an early age at onset with a worse outcome. FNMTC cases Citation: Diquigiovanni, C.; Bonora, E.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • The Concise Guide to Pharmacology 2019/20
    Edinburgh Research Explorer THE CONCISE GUIDE TO PHARMACOLOGY 2019/20 Citation for published version: Cgtp Collaborators 2019, 'THE CONCISE GUIDE TO PHARMACOLOGY 2019/20: Transporters', British Journal of Pharmacology, vol. 176 Suppl 1, pp. S397-S493. https://doi.org/10.1111/bph.14753 Digital Object Identifier (DOI): 10.1111/bph.14753 Link: Link to publication record in Edinburgh Research Explorer Document Version: Publisher's PDF, also known as Version of record Published In: British Journal of Pharmacology General rights Copyright for the publications made accessible via the Edinburgh Research Explorer is retained by the author(s) and / or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Take down policy The University of Edinburgh has made every reasonable effort to ensure that Edinburgh Research Explorer content complies with UK legislation. If you believe that the public display of this file breaches copyright please contact [email protected] providing details, and we will remove access to the work immediately and investigate your claim. Download date: 28. Sep. 2021 S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2019/20: Transporters. British Journal of Pharmacology (2019) 176, S397–S493 THE CONCISE GUIDE TO PHARMACOLOGY 2019/20: Transporters Stephen PH Alexander1 , Eamonn Kelly2, Alistair Mathie3 ,JohnAPeters4 , Emma L Veale3 , Jane F Armstrong5 , Elena Faccenda5 ,SimonDHarding5 ,AdamJPawson5 , Joanna L
    [Show full text]
  • Westminsterresearch ZFP36 Proteins and Mrna Targets in B Cell
    WestminsterResearch http://www.westminster.ac.uk/westminsterresearch ZFP36 proteins and mRNA targets in B cell malignancies Alcaraz, A. This is an electronic version of a PhD thesis awarded by the University of Westminster. © Miss Amor Alcaraz, 2015. The WestminsterResearch online digital archive at the University of Westminster aims to make the research output of the University available to a wider audience. Copyright and Moral Rights remain with the authors and/or copyright owners. Whilst further distribution of specific materials from within this archive is forbidden, you may freely distribute the URL of WestminsterResearch: ((http://westminsterresearch.wmin.ac.uk/). In case of abuse or copyright appearing without permission e-mail [email protected] ZFP36 proteins and mRNA targets in B cell malignancies Maria del Amor Alcaraz-Serrano A Thesis submitted in partial fulfilment of the requirements of the University of Westminster for the degree of Doctor of Philosophy September 2015 Abstract The ZFP36 proteins are a family of post-transcriptional regulator proteins that bind to adenine uridine rich elements (AREs) in 3’ untranslated (3’UTR) regions of mRNAs. The members of the human family, ZFP36L1, ZFP36L2 and ZFP36 are able to degrade mRNAs of important cell regulators that include cytokines, cell signalling proteins and transcriptional factors. This project investigated two proposed targets for the protein family that have important roles in B cell biology, BCL2 and CD38 mRNAs. BCL2 is an anti-apoptotic protein with key roles in cell survival and carcinogenesis; CD38 is a membrane protein differentially expressed in B cells and with a prognostic value in B chronic lymphocytic leukaemia (B-CLL), patients positive for CD38 are considered to have a poor prognosis.
    [Show full text]
  • Polycomb Repressor Complex 2 Function in Breast Cancer (Review)
    INTERNATIONAL JOURNAL OF ONCOLOGY 57: 1085-1094, 2020 Polycomb repressor complex 2 function in breast cancer (Review) COURTNEY J. MARTIN and ROGER A. MOOREHEAD Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G2W1, Canada Received July 10, 2020; Accepted September 7, 2020 DOI: 10.3892/ijo.2020.5122 Abstract. Epigenetic modifications are important contributors 1. Introduction to the regulation of genes within the chromatin. The poly- comb repressive complex 2 (PRC2) is a multi‑subunit protein Epigenetic modifications, including DNA methylation complex that is involved in silencing gene expression through and histone modifications, play an important role in gene the trimethylation of lysine 27 at histone 3 (H3K27me3). The regulation. The dysregulation of these modifications can dysregulation of this modification has been associated with result in pathogenicity, including tumorigenicity. Research tumorigenicity through the increased repression of tumour has indicated an important influence of the trimethylation suppressor genes via condensing DNA to reduce access to the modification at lysine 27 on histone H3 (H3K27me3) within transcription start site (TSS) within tumor suppressor gene chromatin. This methylation is involved in the repression promoters. In the present review, the core proteins of PRC2, as of multiple genes within the genome by condensing DNA well as key accessory proteins, will be described. In addition, to reduce access to the transcription start site (TSS) within mechanisms controlling the recruitment of the PRC2 complex gene promoter sequences (1). The recruitment of H1.2, an H1 to H3K27 will be outlined. Finally, literature identifying the histone subtype, by the H3K27me3 modification has been a role of PRC2 in breast cancer proliferation, apoptosis and suggested as a mechanism for mediating this compaction (1).
    [Show full text]