S4 Were Obtained from BD Pharmingen and R & D Systems

Total Page:16

File Type:pdf, Size:1020Kb

S4 Were Obtained from BD Pharmingen and R & D Systems Supporting Information Billerbeck et al. 10.1073/pnas.0914839107 SI Methods ELISpot. To analyze HCV-specific cytokine–secreting CD8+ T Antibodies and Staining. Anti–CD8-PerCP, anti–CD8-PE, anti– cells, ELISpot assays were performed with intrahepatic and pe- + CD8-PerCP-Cy5.5, anti–CD8-Alexa Fluor 700–anti–CD4-PerCP, ripheral CD8 T cells from chronically HCV-infected patients. + anti–CD4-FITC, anti–CD4-APC, anti–IFN-γ-FITC, anti–TCRγδ- Intrahepatic and peripheral CD8 T cells were isolated and PE, anti–TCRαβ-PE, anti–TNF-α,isotypePE,isotypeFITC,and expanded in an antigen nonspecific way (2 to 4 weeks). IL-17 γ isotypeAPC were obtainedfrom BD Pharmingen. Anti–CD3-FITC/ ELISpot kits (R & D Systems) and IFN- ELISpot kits (Diac- ’ APC, anti–IL-22-APC, anti–CCR6-APC, anti–CCR2-APC, anti– lone) were used according to the manufacturers instructions, × 5 CXCR6-PE, anti–CXCR3-FITC, anti–CCR7-FITC, biotinylated and 1 10 cells were plated per well. Peptides were added at a fi μ polyclonal goat anti–IL-23R, and anti–IFN-γ-FITC were obtained nal concentration of 10 g/mL per peptide and were combined – in pools of 10 peptides, with each peptide being contained in two from R & D Systems; and anti CD161-APC from Miltenyi Biotech. fi Anti–IL-17A-PE, anti–IL-17A-Alexa 647, anti–TCRαβ-APC, anti– different pools in a matrix setup, allowing the identi cation of CD127-PE, anti–RORγT-PE, and anti–IL18R-FITC were from single positive peptides. Tests with peptide pools were per- formed in duplicates; two wells without peptides served as neg- eBioscience. Anti–CD161-PE, anti–CD3-ECD, and anti–TCRγδ- ative controls; four wells each stimulated with PMA/ionomycin FITC were obtained from Immunotech/Beckman Coulter. Anti– served as positive controls. Responses were considered positive if granzyme B-APC and anti–CD3-PacificOrangewereobtainedfrom the number of spots per well was at least three times as high as Caltag Laboratories and anti–Vα24-FITC from Serotec. Anti- – γ the mean of negative controls (typically 0 to 2 spots per well). Runx2 was obtained from Santa Cruz Biotechnology. Anti ROR T- Overlapping peptides derived from HCV strain H77 (genotype Alexa 488 was obtained from Cambridge Biosciences. FITC- or 1a) spanning 18 aa and overlapping by 11 aa were obtained from APC-conjugated antibodies to CD45RA, CD45RO, CD38, CD62L, β Beiresources (National Institutes of Health Biodefense and CD56, CD103, TGF RII, PD-1, ICOS, GITR, CD38, CD62L, Emerging Infections Research Resources Repository). CCR5, CXCR4, CCR9, CD85j, CD27, and CD28 for studies in Fig. − S4 were obtained from BD Pharmingen and R & D Systems. Ex- ELISA. Purified CD161 or CD161+ CD8+ T cells (1 × 105 per well) travidin-PE was obtained from Sigma. were cultured in a 96-well plate and stimulated with PMA/ + For immunofluorescence experiments, CD8 T cells were iso- ionomycin for 48 h. The concentrations of IL-17 and IFN-γ in the lated using an EasySep CD8 negative selection kit and positive PE supernatants were analyzed using human IL-17 or INF-γ Duoset selection beads (Stem Cell Technologies) following anti-IL18R or ELISA (R & D Systems) according to the manufacturer’s in- anti-CD161 staining. Cells were plated in eight-chamber slides structions. coated with CC2 (Nunc), fixed with methanol/acetone, air dried, blocked with 1% BSA, and washed with PBS/Tween 1% solution Microarray Data Analysis. Gene expression profiles for three before staining. For intracellular staining by flow cytometry per- sorted CD8 cell subsets from each of four healthy donors meabilization buffers from BD Biosciences and eBiosciences were were obtained using Illumina Sentrix Human Whole Genome used according to the manufacturer’s instructions. 6 BeadChips. Raw data were exported from the BeadStudio CD8 selection and polyclonal expansion of PBMCs. PBMCs (4 × 106) software (v3.3.7; Illumina) for further processing and analysis wereresuspendedin5mLPBSsolutionandincubatedwithCD8 using the R statistical software and BioConductor packages (1– Dynabeads (Invitrogen) for 20 min at 4 °C. Bound CD4+ Tcells 5). Signal data and detection scores were extracted for each of were isolated by a particle magnetic concentrator. The purity of the 12 samples. Signal data were background corrected using CD4+ Tcellswas>95% by FACS analysis. The CD4+ Tcells array-specific measures of background intensity based on were plated into one well of a 24-well plate (Greiner) in 1 mL negative control probes before being transformed and nor- complete medium (RPMI medium 1640 containing 10% FCS, malized using the vs.n2 BioConductor package. After filtering out probes not detected in any of the 12 samples 1% streptomycin/penicillin, and 1.5% 1 M Hepes containing 100 < U/mL IL-2; Hoffmann-La Roche), 0.04 g/mL anti–human CD3 (detection score 0.95), 32,159 probes were retained for further monoclonal antibody (Immunotech), and 2 × 106 irradiated analysis. Statistical testing was performed using the Linear Models for autologous PBMCs as feeder cells. Twice per week, 1 mL of Microarray Analysis (limma) BioConductor package. Three con- medium was exchanged and 100 U/mL IL-2 was added. After 3 + trasts were generated corresponding to the possible cell type com- weeks, the expanded CD8 PBMCs and intrahepatic lympho- − parisons (CD161++ vs. CD161+, CD161++ vs. CD161 ,CD161+ cytes were analyzed. − vs. CD161 ), each using a paired design. Raw P values were cor- CD8 selection and polyclonal expansion of LILs. Briefly, the liver biopsy rected for multiple testing using the false discovery rate controlling specimens were homogenized using a 70-m Dounce tissue grinder procedure of Benjamini and Hochberg. Gene details were added (BD Biosciences). Cell suspensions were incubated with CD8 fi fi + to nal probe lists using the relevant annotation le (HumanWG- Dynabeads (Invitrogen) for 20 min at 4 °C. Bound CD8 cells were 6_V3_0_R0_11282955_A.txt) downloaded from the Illumina Web isolated using a particle magnetic concentrator. The purity of site (http://www.illumina.com). CD8+ cells was >95% by FACS analysis. The intrahepatic CD4+ T + cells were then expanded as described earlier for CD8 PBMCs. RNA Isolation, cDNA Synthesis, and Quantitative RT-PCR. RNA was + Importantly, the expansion of peripheral and intrahepatic CD8 T isolated from sorted cells using the miRNeasy Mini Kit (Qia- cells from a given patient was always performed in parallel and for gen). cDNA synthesis was performed using a mixture of random the same time before analysis to obtain comparable results for and oligo(dT) primers and SuperScript III reverse transcriptase both compartments. Tonsil/kidney–derived lymphocytes were (Invitogen). Quantitative real-time PCR reactions were carried prepared similarly, although without CD8+ T cell selection and out using a LightCycler 480 (Roche Diagnostics) and a com- culture, and joint-derived lymphocytes prepared without the ho- bination of preoptimized Qiagen QuantiTect Primer Assays mogenization step. [IL-23R (Hs_IL23R_1_SG) and HPRT1 as an internal control Billerbeck et al. www.pnas.org/cgi/content/short/0914839107 1of14 (Hs_HPRT1_1_SG)] and primers designed using Primer36 (RORc, manufacturer’s instructions, with cycling conditions as follows: 5′-AGAAGGACAGGGAGCCAAG-3′ for forward and 5′-CAA- 95 °C for 15 min and 40 cycles of 94 °C for 15 s, 54 °C for 30 s, and GGGATCACTTCAATTTGTG-3′ for reverse). LightCycler 480 72 °C for 30 s. Fold expression changes were determined by the 2- SYBR Green I Master Mix (Roche) was used according to the ΔΔCT method (7). 1. R Development Core Team (2007) R: a language and environment for statistical com- 5. Benjamini Y, Hochberg Y (1995) Controlling the false discovery rate: a practical and puting (R Foundation, Vienna). powerful approach to multiple testing. J R Stat Soc, B 57:289–300. 2. Gentleman RC, et al. (2004) Bioconductor: open software development for computational 6. Rozen S, Skaletsky H (2000) Primer3 on the WWW for general users and for biology and bioinformatics. Genome Biol 5:R80. biologist programmers. In Bioinformatics methods and protocols: methods in 3. Huber W, von Heydebreck A, Sültmann H, Poustka A, Vingron M (2002) Variance molecular biology, eds Krawetz S, Misener S (Humana Press, Totowa, NJ), pp stabilization applied to microarray data calibration and to the quantification of 365–386. differential expression. Bioinformatics 18 (suppl 1):S96–S104. 7. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using 4. Smyth GK (2005) Limma: linear models for microarray data. In Bioinformatics and real-time quantitative PCR and the 2(-delta delta C(T)) method. Methods 25: computational biology solutions using R and Bioconductor, ed Gentleman R (Springer- – Verlag, New York), pp 397–420. 402 408. 29.1 2.37 12.3 0.72 5 A 105 10 8.58 4 104 10 3 103 10 CD8 <FITC-A> IFN- <APC-A> CD161 2 102 10 0 0 CD161++gate 86.5 0.47 62.1 6.45 2 3 4 5 0102 103 104 105 010 10 10 10 <PE-A> <PE-A> B 7.64 0.57 27.7 1.42 5 10 105 4 8.13 10 104 3 10 103 CD4 <APC-A> <FITC-A> IFN- CD161 2 2 10 10 CD161+gate 0 0 91.5 0.3 65.1 5.83 2 3 4 5 010 10 10 10 0102 103 104 105 <PE-A> <PE-A> IL-17 IL-17 C 10.0 p<0.0001 10.0 p=n.s. 7.5 7.5 CD4 5.0 5.0 2.5 2.5 % IL-17+ T cells % IL-17+ T cells 0.0 0.0 CD161+ CD161- CD8 CD4 D 2500 200 200 CD161++ CD161+ CD161- 2000 150 150 1500 0.2 0.9 0.41 100 Unstim. # Cells # Cells 100 # Cells 1000 50 50 500 0 0 0 0 1 2 3 4 100 101 102 103 104 100 101 102 103 104 10 10 10 10 10 FL1-H: IFN-G FITC FL1-H: IFN-G FITC FL1-H: IFN-G FITC Fig.
Recommended publications
  • Killer-Like Receptors and GPR56 Progressive Expression Defines Cytokine Production of Human CD4+ Memory T Cells
    ARTICLE https://doi.org/10.1038/s41467-019-10018-1 OPEN Killer-like receptors and GPR56 progressive expression defines cytokine production of human CD4+ memory T cells Kim-Long Truong1,7, Stephan Schlickeiser1,2,7, Katrin Vogt1, David Boës1, Katarina Stanko1, Christine Appelt1, Mathias Streitz1, Gerald Grütz1,2, Nadja Stobutzki1, Christian Meisel1, Christina Iwert1, Stefan Tomiuk3, Julia K. Polansky2,4, Andreas Pascher5, Nina Babel2,6, Ulrik Stervbo 6, Igor Sauer 5, Undine Gerlach5 & Birgit Sawitzki1,2 1234567890():,; All memory T cells mount an accelerated response on antigen reencounter, but significant functional heterogeneity is present within the respective memory T-cell subsets as defined by CCR7 and CD45RA expression, thereby warranting further stratification. Here we show that several surface markers, including KLRB1, KLRG1, GPR56, and KLRF1, help define low, high, or exhausted cytokine producers within human peripheral and intrahepatic CD4+ memory T-cell populations. Highest simultaneous production of TNF and IFN-γ is observed in KLRB1+KLRG1+GPR56+ CD4 T cells. By contrast, KLRF1 expression is associated with T-cell exhaustion and reduced TNF/IFN-γ production. Lastly, TCRβ repertoire analysis and in vitro differentiation support a regulated, progressive expression for these markers during CD4+ memory T-cell differentiation. Our results thus help refine the classification of human memory T cells to provide insights on inflammatory disease progression and immunotherapy development. 1 Institute of Medical Immunology, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 13353 Berlin, Germany. 2 Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité – Universitätsmedizin Berlin, 13353 Berlin, Germany. 3 Milteny Biotec GmbH, 51429 Bergisch Gladbach, Germany.
    [Show full text]
  • NK Cell Memory to Cytomegalovirus: Implications for Vaccine Development
    Review NK Cell Memory to Cytomegalovirus: Implications for Vaccine Development Calum Forrest 1 , Ariane Gomes 1 , Matthew Reeves 1,* and Victoria Male 2,* 1 Institute of Immunity & Transplantation, UCL, Royal Free Campus, London NW3 2PF, UK; [email protected] (C.F.); [email protected] (A.G.) 2 Department of Metabolism, Digestion and Reproduction, Imperial College London, Chelsea and Westminster Campus, London SW10 9NH, UK * Correspondence: [email protected] (M.R.); [email protected] (V.M.) Received: 24 June 2020; Accepted: 15 July 2020; Published: 20 July 2020 Abstract: Natural killer (NK) cells are innate lymphoid cells that recognize and eliminate virally-infected and cancerous cells. Members of the innate immune system are not usually considered to mediate immune memory, but over the past decade evidence has emerged that NK cells can do this in several contexts. Of these, the best understood and most widely accepted is the response to cytomegaloviruses, with strong evidence for memory to murine cytomegalovirus (MCMV) and several lines of evidence suggesting that the same is likely to be true of human cytomegalovirus (HCMV). The importance of NK cells in the context of HCMV infection is underscored by the armory of NK immune evasion genes encoded by HCMV aimed at subverting the NK cell immune response. As such, ongoing studies that have utilized HCMV to investigate NK cell diversity and function have proven instructive. Here, we discuss our current understanding of NK cell memory to viral infection with a focus on the response to cytomegaloviruses. We will then discuss the implications that this will have for the development of a vaccine against HCMV with particular emphasis on how a strategy that can harness the innate immune system and NK cells could be crucial for the development of a vaccine against this high-priority pathogen.
    [Show full text]
  • Anti-LYN Monoclonal Antibody, Clone 2285DU33.4.2.2 (DCABY-1218) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use
    Anti-LYN monoclonal antibody, clone 2285DU33.4.2.2 (DCABY-1218) This product is for research use only and is not intended for diagnostic use. PRODUCT INFORMATION Antigen Description Non-receptor tyrosine-protein kinase that transmits signals from cell surface receptors and plays an important role in the regulation of innate and adaptive immune responses, hematopoiesis, responses to growth factors and cytokines, integrin signaling, but also responses to DNA damage and genotoxic agents. Functions primarily as negative regulator, but can also function as activator, depending on the context. Required for the initiation of the B-cell response, but also for its down- regulation and termination. Plays an important role in the regulation of B-cell differentiation, proliferation, survival and apoptosis, and is important for immune self-tolerance. Acts downstream of several immune receptors, including the B-cell receptor, CD79A, CD79B, CD5, CD19, CD22, FCER1, FCGR2, FCGR1A, TLR2 and TLR4. Plays a role in the inflammatory response to bacterial lipopolysaccharide. Mediates the responses to cytokines and growth factors in hematopoietic progenitors, platelets, erythrocytes, and in mature myeloid cells, such as dendritic cells, neutrophils and eosinophils. Acts downstream of EPOR, KIT, MPL, the chemokine receptor CXCR4, as well as the receptors for IL3, IL5 and CSF2. Plays an important role in integrin signaling. Regulates cell proliferation, survival, differentiation, migration, adhesion, degranulation, and cytokine release. Down- regulates signaling pathways by phosphorylation of immunoreceptor tyrosine-based inhibitory motifs (ITIM), that then serve as binding sites for phosphatases, such as PTPN6/SHP-1, PTPN11/SHP-2 and INPP5D/SHIP-1, that modulate signaling by dephosphorylation of kinases and their substrates.
    [Show full text]
  • Genitourinary Pathology (Including Renal Tumors)
    LABORATORY INVESTIGATION THE BASIC AND TRANSLATIONAL PATHOLOGY RESEARCH JOURNAL LI VOLUME 99 | SUPPLEMENT 1 | MARCH 2019 2019 ABSTRACTS GENITOURINARY PATHOLOGY (INCLUDING RENAL TUMORS) (776-992) MARCH 16-21, 2019 PLATF OR M & 2 01 9 ABSTRACTS P OSTER PRESENTATI ONS EDUCATI ON C O M MITTEE Jason L. Hornick , C h air Ja mes R. Cook R h o n d a K. Y a nti s s, Chair, Abstract Revie w Board S ar a h M. Dr y and Assign ment Co m mittee Willi a m C. F a q ui n Laura W. La mps , Chair, C ME Subco m mittee C ar ol F. F ar v er St e v e n D. Billi n g s , Interactive Microscopy Subco m mittee Y uri F e d ori w Shree G. Shar ma , Infor matics Subco m mittee Meera R. Ha meed R aj a R. S e et h al a , Short Course Coordinator Mi c h ell e S. Hir s c h Il a n W ei nr e b , Subco m mittee for Unique Live Course Offerings Laksh mi Priya Kunju D a vi d B. K a mi n s k y ( Ex- Of ici o) A n n a M ari e M ulli g a n Aleodor ( Doru) Andea Ri s h P ai Zubair Baloch Vi nita Parkas h Olca Bast urk A nil P ar w a ni Gregory R. Bean , Pat h ol o gist-i n- Trai ni n g D e e p a P atil D a ni el J.
    [Show full text]
  • 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,
    [Show full text]
  • Monoclonal Antibody to CD161 / KLRB1 - Purified
    OriGene Technologies, Inc. OriGene Technologies GmbH 9620 Medical Center Drive, Ste 200 Schillerstr. 5 Rockville, MD 20850 32052 Herford UNITED STATES GERMANY Phone: +1-888-267-4436 Phone: +49-5221-34606-0 Fax: +1-301-340-8606 Fax: +49-5221-34606-11 [email protected] [email protected] SM1611PT Monoclonal Antibody to CD161 / KLRB1 - Purified Alternate names: C-type lectin domain family 5 member B, CLEC5B, HNKR-P1a, Killer cell lectin-like receptor subfamily B member 1, NKRP1A, Natural killer cell surface protein P1A Quantity: 25 µg Concentration: 1.0 mg/ml Background: Natural killer (NK) cells are lymphocytes that mediate cytotoxicity and secrete cytokines after immune stimulation. Several genes of the C-type lectin superfamily, including the rodent NKRP1 family of glycoproteins, are expressed by NK cells and may be involved in the regulation of NK cell function. The KLRB1 (CD161) protein contains an extracellular domain with several motifs characteristic of C type lectins, a transmembrane domain, and a cytoplasmic domain. The KLRB1 protein is classified as a type II membrane protein because it has an external C terminus. In mouse the NKRP1 family has three members, NKRP1A, B and C, whilst in human only one member has been identified. The human protein has received the designation CD161, and the mouse proteins have been referred to as CD161a, b and c. Engagement of CD161c has been reported to have activating function in NK cells, whilst engagement of CD161b is inhibitory. CD161 is expressed by almost all NK cells and with a small subset of CD3+ve T cells.
    [Show full text]
  • CD29 Identifies IFN-Γ–Producing Human CD8+ T Cells With
    + CD29 identifies IFN-γ–producing human CD8 T cells with an increased cytotoxic potential Benoît P. Nicoleta,b, Aurélie Guislaina,b, Floris P. J. van Alphenc, Raquel Gomez-Eerlandd, Ton N. M. Schumacherd, Maartje van den Biggelaarc,e, and Monika C. Wolkersa,b,1 aDepartment of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; bLandsteiner Laboratory, Oncode Institute, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; cDepartment of Research Facilities, Sanquin Research, 1066 CX Amsterdam, The Netherlands; dDivision of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; and eDepartment of Molecular and Cellular Haemostasis, Sanquin Research, 1066 CX Amsterdam, The Netherlands Edited by Anjana Rao, La Jolla Institute for Allergy and Immunology, La Jolla, CA, and approved February 12, 2020 (received for review August 12, 2019) Cytotoxic CD8+ T cells can effectively kill target cells by producing therefore developed a protocol that allowed for efficient iso- cytokines, chemokines, and granzymes. Expression of these effector lation of RNA and protein from fluorescence-activated cell molecules is however highly divergent, and tools that identify and sorting (FACS)-sorted fixed T cells after intracellular cytokine + preselect CD8 T cells with a cytotoxic expression profile are lacking. staining. With this top-down approach, we performed an un- + Human CD8 T cells can be divided into IFN-γ– and IL-2–producing biased RNA-sequencing (RNA-seq) and mass spectrometry cells. Unbiased transcriptomics and proteomics analysis on cytokine- γ– – + + (MS) analyses on IFN- and IL-2 producing primary human producing fixed CD8 T cells revealed that IL-2 cells produce helper + + + CD8 Tcells.
    [Show full text]
  • Characteristics of B Cell-Associated Gene Expression in Patients With
    MOLECULAR MEDICINE REPORTS 13: 4113-4121, 2016 Characteristics of B cell-associated gene expression in patients with coronary artery disease WENWEN YAN*, HAOMING SONG*, JINFA JIANG, WENJUN XU, ZHU GONG, QIANGLIN DUAN, CHUANGRONG LI, YUAN XIE and LEMIN WANG Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China Received May 19, 2015; Accepted February 12, 2016 DOI: 10.3892/mmr.2016.5029 Abstract. The current study aimed to identify differentially with the two other groups. Additionally the gene expression expressed B cell-associated genes in peripheral blood mono- levels of B cell regulatory genes were measured. In patients nuclear cells and observe the changes in B cell activation at with AMI, CR1, LILRB2, LILRB3 and VAV1 mRNA expres- different stages of coronary artery disease. Groups of patients sion levels were statistically increased, whereas, CS1 and IL4I1 with acute myocardial infarction (AMI) and stable angina (SA), mRNAs were significantly reduced compared with the SA and as well as healthy volunteers, were recruited into the study control groups. There was no statistically significant difference (n=20 per group). Whole human genome microarray analysis in B cell-associated gene expression levels between patients was performed to examine the expression of B cell-associated with SA and the control group. The present study identified the genes among these three groups. The mRNA expression levels downregulation of genes associated with BCRs, B2 cells and of 60 genes associated with B cell activity and regulation were B cell regulators in patients with AMI, indicating a weakened measured using reverse transcription-quantitative polymerase T cell-B cell interaction and reduced B2 cell activation during chain reaction.
    [Show full text]
  • A Novel CD4+ CTL Subtype Characterized by Chemotaxis and Inflammation Is Involved in the Pathogenesis of Graves’ Orbitopa
    Cellular & Molecular Immunology www.nature.com/cmi ARTICLE OPEN A novel CD4+ CTL subtype characterized by chemotaxis and inflammation is involved in the pathogenesis of Graves’ orbitopathy Yue Wang1,2,3,4, Ziyi Chen 1, Tingjie Wang1,2, Hui Guo1, Yufeng Liu2,3,5, Ningxin Dang3, Shiqian Hu1, Liping Wu1, Chengsheng Zhang4,6,KaiYe2,3,7 and Bingyin Shi1 Graves’ orbitopathy (GO), the most severe manifestation of Graves’ hyperthyroidism (GH), is an autoimmune-mediated inflammatory disorder, and treatments often exhibit a low efficacy. CD4+ T cells have been reported to play vital roles in GO progression. To explore the pathogenic CD4+ T cell types that drive GO progression, we applied single-cell RNA sequencing (scRNA-Seq), T cell receptor sequencing (TCR-Seq), flow cytometry, immunofluorescence and mixed lymphocyte reaction (MLR) assays to evaluate CD4+ T cells from GO and GH patients. scRNA-Seq revealed the novel GO-specific cell type CD4+ cytotoxic T lymphocytes (CTLs), which are characterized by chemotactic and inflammatory features. The clonal expansion of this CD4+ CTL population, as demonstrated by TCR-Seq, along with their strong cytotoxic response to autoantigens, localization in orbital sites, and potential relationship with disease relapse provide strong evidence for the pathogenic roles of GZMB and IFN-γ-secreting CD4+ CTLs in GO. Therefore, cytotoxic pathways may become potential therapeutic targets for GO. 1234567890();,: Keywords: Graves’ orbitopathy; single-cell RNA sequencing; CD4+ cytotoxic T lymphocytes Cellular & Molecular Immunology
    [Show full text]
  • Supplementary Material DNA Methylation in Inflammatory Pathways Modifies the Association Between BMI and Adult-Onset Non- Atopic
    Supplementary Material DNA Methylation in Inflammatory Pathways Modifies the Association between BMI and Adult-Onset Non- Atopic Asthma Ayoung Jeong 1,2, Medea Imboden 1,2, Akram Ghantous 3, Alexei Novoloaca 3, Anne-Elie Carsin 4,5,6, Manolis Kogevinas 4,5,6, Christian Schindler 1,2, Gianfranco Lovison 7, Zdenko Herceg 3, Cyrille Cuenin 3, Roel Vermeulen 8, Deborah Jarvis 9, André F. S. Amaral 9, Florian Kronenberg 10, Paolo Vineis 11,12 and Nicole Probst-Hensch 1,2,* 1 Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland; [email protected] (A.J.); [email protected] (M.I.); [email protected] (C.S.) 2 Department of Public Health, University of Basel, 4001 Basel, Switzerland 3 International Agency for Research on Cancer, 69372 Lyon, France; [email protected] (A.G.); [email protected] (A.N.); [email protected] (Z.H.); [email protected] (C.C.) 4 ISGlobal, Barcelona Institute for Global Health, 08003 Barcelona, Spain; [email protected] (A.-E.C.); [email protected] (M.K.) 5 Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain 6 CIBER Epidemiología y Salud Pública (CIBERESP), 08005 Barcelona, Spain 7 Department of Economics, Business and Statistics, University of Palermo, 90128 Palermo, Italy; [email protected] 8 Environmental Epidemiology Division, Utrecht University, Institute for Risk Assessment Sciences, 3584CM Utrecht, Netherlands; [email protected] 9 Population Health and Occupational Disease, National Heart and Lung Institute, Imperial College, SW3 6LR London, UK; [email protected] (D.J.); [email protected] (A.F.S.A.) 10 Division of Genetic Epidemiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; [email protected] 11 MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, W2 1PG London, UK; [email protected] 12 Italian Institute for Genomic Medicine (IIGM), 10126 Turin, Italy * Correspondence: [email protected]; Tel.: +41-61-284-8378 Int.
    [Show full text]
  • Single-Cell Analysis of Crohn's Disease Lesions Identifies
    bioRxiv preprint doi: https://doi.org/10.1101/503102; this version posted December 20, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Single-cell analysis of Crohn’s disease lesions identifies a pathogenic cellular module associated with resistance to anti-TNF therapy JC Martin1,2,3, G Boschetti1,2,3, C Chang1,2,3, R Ungaro4, M Giri5, LS Chuang5, S Nayar5, A Greenstein6, M. Dubinsky7, L Walker1,2,5,8, A Leader1,2,3, JS Fine9, CE Whitehurst9, L Mbow9, S Kugathasan10, L.A. Denson11, J.Hyams12, JR Friedman13, P Desai13, HM Ko14, I Laface1,2,8, Guray Akturk1,2,8, EE Schadt15,16, S Gnjatic1,2,8, A Rahman1,2,5,8, , M Merad1,2,3,8,17,18*, JH Cho5,17,*, E Kenigsberg1,15,16,17* 1 Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA. 2 Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA. 3 Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA. 4 The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA. 5 Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA. 6 Department of Colorectal Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA 7 Department of Pediatrics, Susan and Leonard Feinstein IBD Clinical Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
    [Show full text]
  • High-Dimensional Analysis Reveals a Distinct Population of Adaptive-Like Tissue
    bioRxiv preprint doi: https://doi.org/10.1101/2019.12.20.883785; this version posted December 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 High-dimensional analysis reveals a distinct population of adaptive-like tissue- 2 resident NK cells in human lung 3 4 Nicole Marquardt1*, Marlena Scharenberg1, Jeffrey E. Mold2, Joanna Hård2, Eliisa 5 Kekäläinen3,4, Marcus Buggert1, Son Nguyen5,6, Jennifer N. Wilson1, Mamdoh Al- 6 Ameri7, Hans-Gustaf Ljunggren1, Jakob Michaëlsson1 7 8 Running title: Adaptive-like human lung tissue-resident NK cells 9 10 Affiliations: 11 1Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, 12 Stockholm, Sweden 13 2Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden 14 3Translational Immunology Research Program & Department of Bacteriology and 15 Immunology, University of Helsinki, Finland 16 4HUSLAB, Division of Clinical Microbiology, Helsinki University Hospital, Helsinki, 17 Finland, 18 5Department of Microbiology, Perelman School of Medicine, University of 19 Pennsylvania, Philadelphia, PA, USA 20 6Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, 21 Philadelphia, PA, USA 22 7Thoracic Surgery, Department of Molecular Medicine and Surgery, Karolinska 23 Institutet, Karolinska University Hospital, Stockholm, Sweden 24 1 bioRxiv preprint doi: https://doi.org/10.1101/2019.12.20.883785; this version posted December 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]