Analysis of the Immune Responses in the Ileum of Gnotobiotic Pigs Infected with the Recombinant GII.P12 GII.3 Human Norovirus by Mrna Sequencing

Total Page:16

File Type:pdf, Size:1020Kb

Analysis of the Immune Responses in the Ileum of Gnotobiotic Pigs Infected with the Recombinant GII.P12 GII.3 Human Norovirus by Mrna Sequencing viruses Article Analysis of the Immune Responses in the Ileum of Gnotobiotic Pigs Infected with the Recombinant GII.p12_GII.3 Human Norovirus by mRNA Sequencing Byung-Joo Park 1, Hee-Seop Ahn 1, Sang-Hoon Han 1, Hyeon-Jeong Go 1, Dong-Hwi Kim 1, Changsun Choi 2 , Soontag Jung 2 , Jinjong Myoung 3, Joong-Bok Lee 1, Seung-Yong Park 1, Chang-Seon Song 1 , Sang-Won Lee 1, Hoon-Taek Lee 4 and In-Soo Choi 1,* 1 Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea; [email protected] (B.-J.P.); [email protected] (H.-S.A.); [email protected] (S.-H.H.); [email protected] (H.-J.G.); [email protected] (D.-H.K.); [email protected] (J.-B.L.); [email protected] (S.-Y.P.); [email protected] (C.-S.S.); [email protected] (S.-W.L.) 2 Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Gyeonggi 17546, Korea; [email protected] (C.C.); [email protected] (S.J.) 3 Korea Zoonosis Research Institute, Chonbuk National University, Jeonju, Jeollabuk-do 54896, Korea; [email protected] 4 Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea; [email protected] * Correspondence: [email protected]; Tel.: +82-2049-6228 Abstract: Norovirus genogroup II (NoV GII) induces acute gastrointestinal food-borne illness in humans. Because gnotobiotic pigs can be infected with human norovirus (HuNoV) GII, they are frequently used to analyze the associated pathogenic mechanisms and immune responses, which Citation: Park, B.-J.; Ahn, H.-S.; Han, remain poorly understood. Recently, mRNA sequencing analysis (RNA-Seq) has been used to S.-H.; Go, H.-J.; Kim, D.-H.; Choi, C.; identify cellular responses to viruses. In this study, we investigated the host immune response Jung, S.; Myoung, J.; Lee, J.-B.; Park, S.-Y.; et al. Analysis of the Immune and possible mechanisms involved in virus evasion in the ileum of gnotobiotic pigs infected with Responses in the Ileum of Gnotobiotic HuNoV by RNA-Seq. HuNoV was detected in the feces, blood, and tissues of the jejunum, ileum, Pigs Infected with the Recombinant colon, mesenteric lymph node, and spleen of pigs infected with HuNoV. In analysis of mRNA GII.p12_GII.3 Human Norovirus by sequencing, expression of anti-viral protein genes such as OAS1, MX1, and MX2 were largely mRNA Sequencing. Viruses 2021, 13, decreased, whereas type I IFN was increased in pigs infected with HuNoV. In addition, expression of 92. https://doi.org/10.3390/ TNF and associated anti-inflammatory cytokine genes such as IL10 was increased in HuNoV-infected v13010092 pigs. Expression of genes related to natural killer (NK) cell cytotoxicity and CD8+ T cell exhaustion was increased, whereas that of MHC class I genes was decreased. Expression profiles of selected Academic Editor: Lennart Svensson genes were further confirmed by qRT-PCR and Western blot. These results suggest that infection with Received: 7 December 2020 HuNoV induces NK cell-mediated cytotoxicity but suppresses type I IFN- and CD8+ T cell-mediated Accepted: 8 January 2021 antiviral responses. Published: 11 January 2021 Keywords: human norovirus; gnotobiotic pig; immune response; high-throughput mRNA sequencing Publisher’s Note: MDPI stays neu- tral with regard to jurisdictional clai- ms in published maps and institutio- nal affiliations. 1. Introduction Norovirus (NoV) constitutes an important pathogen of food-borne illnesses in humans. Human norovirus (HuNoV) is transmitted by the fecal–oral route and generally causes Copyright: © 2021 by the authors. Li- self-limiting acute gastroenteritis characterized by diarrhea, vomiting, stomach pain, and censee MDPI, Basel, Switzerland. fever [1]. HuNoV infection leads to approximately 20 million illnesses, 60,000 hospitaliza- This article is an open access article tions, and 700 deaths annually in the United States [2,3]. NoVs comprise non-enveloped distributed under the terms and con- single-stranded positive-sense RNA viruses belonging to the family Caliciviridae [1]. They ditions of the Creative Commons At- tribution (CC BY) license (https:// are putatively classified into ten genogroups and further divided into at least 49 geno- creativecommons.org/licenses/by/ types [4], with NoVs of genogroups I (GI), II (GII), and IV (GIV) representing the main 4.0/). causative agents of most human infections [5]. Viruses 2021, 13, 92. https://doi.org/10.3390/v13010092 https://www.mdpi.com/journal/viruses Viruses 2021, 13, 92 2 of 21 However, although the clinical features of NoV gastroenteritis are well established, the target cells of HuNoV infection and associated host immune responses are poorly understood because of previous difficulties in identifying suitable cell culture systems and establishing animal models [6]. Initially, human B cells were proposed to constitute a major target of HuNoV, with reproductive viral replication being demonstrated in these cells [7]. Human enterocytes, macrophages, T cells, and dendritic cells (DCs) of the small intestine have also been suggested as HuNoV targets [8]. In addition, the production of T helper type 1 (Th1) cytokines and acute chemokines is observed in patients infected with HuNoV [9]. Gnotobiotic pigs have been used as an alternative animal model to study the viral pathogenesis and immune responses to HuNoV infection because they harbor the histo- blood group of antigens shown to function as the receptor for the virus [10]. Newborn gnotobiotic pigs infected with HuNoV demonstrate mild diarrhea, virus replication, and histopathologic lesions in the small intestinal tract [11]. Post-weaning gnotobiotic pigs are also used to study viral pathogenesis of HuNoV infection and target tissues of HuNoV in the pig [12,13]. Those studies identified experimental infectious doses 50 of HuNoV, such as 2.74 × 103 and 6.43 × 104 genomic equivalent (GE) copies in neonatal piglets and 33–34 days old pigs, respectively. They also found that the administration of simvastatin into pigs produced the increased infectivity of HuNoV. Notably, gnotobiotic pigs infected with HuNoV produce high levels of Th1 cytokines, albeit low levels of Th2 and proinflammatory cytokines, similar to that observed in human patients [9,14]. Additionally, a longer period of viral shedding is exhibited in RAG2/IL2RG-deficient pigs, indicating the importance of lymphocyte-mediated adaptive immune responses for elimination of HuNoV [15]. Understanding of NoV viral pathogenesis and immune responses has been expanded since the discovery of murine norovirus (MuNoV) [16]. Whereas HuNoV only induces acute infection in immunocompetent individuals, different MuNoV strains may cause persistent or acute infection in wild-type mice [17,18]. MuNoV induces systemic infec- tion, which leads to dissemination of the virus to various tissues, such as the intestine, spleen, liver, and lung [19]. Moreover, infection by MuNoV has been identified in intestinal macrophages, DCs, B lymphocytes, and T lymphocytes as well [16,20–22]. Although wild- type mice infected with MNV do not experience diarrhea, severe diarrhea is induced in interferon-deficient mice [19]. MNV infection is also controlled by the innate and adaptive immune systems [23–25]. Recently, a gene expression profiling study was conducted via a high-throughput mRNA sequencing (RNA-Seq) method to determine up- and downreg- ulated genes in MuNoV-infected mouse macrophage cell lines [26]. However, it remains unclear how the innate and adaptive immune systems are induced or suppressed in the small intestine during the acute period of HuNoV or MuNoV infection. In this study, we ascertained the comprehensive immune responses induced in the small intestine of gnotobiotic pigs that had been infected with HuNoV. The mRNA expres- sion profiles of the ileum were investigated by using high-throughput sequencing and confirmed by quantitative reverse-transcription-PCR (qRT-PCR) and Western blotting anal- yses. Generally, infection with HuNoV appeared to induce the activation of natural killer (NK) cells and suppression of CD8+ T lymphocytes. This new information, provided in this study, will substantively contribute to an improved understanding of NoV pathogenesis and immunology. 2. Materials and Methods 2.1. Virus Preparation HuNoV strain CAU140599 (GenBank accession no. MN199033), HNV GII.p12_GII.3, obtained from a patient suffering from acute gastroenteritis, was used as the infectious agent [27,28]. The virus stock was diluted 10 fold with minimum essential medium (Wellgene, Gyeongsan, Korea). The diluted solutions were centrifuged at 3000× g for 30 min and the supernatant was collected. RNA was extracted from the supernatant using Viruses 2021, 13, 92 3 of 21 a QIAamp viral RNA extraction kit (Qiagen, Valencia, CA, USA), and the viral RNA was stored at −70 ◦C. 2.2. Animals and Experimental Design A total of 15, four-week-old post-weaning gnotobiotic pigs were provided by the Bio-organ Research Center, Konkuk University, Seoul, Korea. The experimental procedures were approved by the Institutional Animal Care and Use Committee (IACUC), Konkuk University, Korea (approval number KU14073). The pigs were acclimatized for 3 days at a biosecurity level 2 grade facility by providing commercial feed prior to conducting experiments. All infection experiments were performed at the biosecurity level 2 grade- facility located at the College of Veterinary Medicine, Konkuk University. Pigs were divided into two experimental groups: a mock-infected negative control group and a HuNoV-infected group. Five pigs were used as negative controls. Ten pigs were infected with 1 × 107 genome equivalent (GE) copies of HuNoV. Blood and rectal swab samples were collected every day for 3 days after viral infection. All pigs were euthanized at day 3 with potassium chloride after administering zolazepam. Gross pathogenic legions of the small and large intestines, mesenteric lymph nodes, and spleen were examined. The collected organs were stored at –70 ◦C and fixed in 4% formaldehyde. 2.3. Preparation of Fecal and Serum Samples The rectal swab samples collected from the gnotobiotic pigs were diluted 1:10 (w/v) with phosphate-buffered saline (PBS).
Recommended publications
  • Enhancing a Natural Killer: Modification of NK Cells for Cancer
    cells Review Enhancing a Natural Killer: Modification of NK Cells for Cancer Immunotherapy Rasa Islam 1,2, Aleta Pupovac 1, Vera Evtimov 1, Nicholas Boyd 1, Runzhe Shu 1, Richard Boyd 1 and Alan Trounson 1,2,* 1 Cartherics Pty Ltd., Clayton 3168, Australia; [email protected] (R.I.); [email protected] (A.P.); [email protected] (V.E.); [email protected] (N.B.); [email protected] (R.S.); [email protected] (R.B.) 2 Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia * Correspondence: [email protected] Abstract: Natural killer (NK) cells are potent innate immune system effector lymphocytes armed with multiple mechanisms for killing cancer cells. Given the dynamic roles of NK cells in tumor surveillance, they are fast becoming a next-generation tool for adoptive immunotherapy. Many strategies are being employed to increase their number and improve their ability to overcome cancer resistance and the immunosuppressive tumor microenvironment. These include the use of cytokines and synthetic compounds to bolster propagation and killing capacity, targeting immune-function checkpoints, addition of chimeric antigen receptors (CARs) to provide cancer specificity and genetic ablation of inhibitory molecules. The next generation of NK cell products will ideally be readily available as an “off-the-shelf” product and stem cell derived to enable potentially unlimited supply. However, several considerations regarding NK cell source, genetic modification and scale up first Citation: Islam, R.; Pupovac, A.; need addressing. Understanding NK cell biology and interaction within specific tumor contexts Evtimov, V.; Boyd, N.; Shu, R.; Boyd, will help identify necessary NK cell modifications and relevant choice of NK cell source.
    [Show full text]
  • And NK-Cell Neoplasms
    J Hematopathol (2009) 2:65–73 DOI 10.1007/s12308-009-0034-z COMMENT Commentary on the 2008 WHO classification of mature T- and NK-cell neoplasms Megan S. Lim & Laurence de Leval & Leticia Quintanilla-Martinez Received: 29 April 2009 /Accepted: 1 May 2009 /Published online: 27 June 2009 # Springer-Verlag 2009 Abstract In 2008, the World Health Organization (WHO) Introduction published a revised and updated edition of the classification of tumors of the hematopoietic and lymphoid tissues. The The 2008 World Health Organization (WHO) classification aims of the fourth edition of the WHO classification was to of mature thymus (T)- and natural killer (NK)-cell neo- incorporate new scientific and clinical information in order plasms provide clarification regarding diagnostic criteria, to refine diagnostic criteria for previously described neo- etiologic insights, and prognostic information for several plasms and to introduce newly recognized disease entities. new/provisional entities [1]. The recognition that T-cell The recognition that T-cell lymphomas are related to the lymphomas are related to the innate and adaptive immune innate and adaptive immune system, as well as enhanced system, as well as enhanced understanding of other T-cell understanding of other T-cell subsets, such as the regula- subsets, such as the regulatory T-cell and follicular helper tory T-cell and follicular helper T-cells, has contributed to T-cells (TFH), has contributed to our understanding of the our understanding of the morphologic, histologic, and morphologic, histologic, and immunophenotypic features of immunophenotypic features of T- and NK-cell neoplasms. T- and NK-cell neoplasms. New insights regarding immu- The purpose of this review is to highlight major changes in nophenotypic features of large granular lymphocytes and the classification of T- and NK-cell neoplasms and to NK-cells have provided better flow cytometric tools for the explain the rationale for these changes.
    [Show full text]
  • Orderly and Nonstochastic Acquisition of CD94/NKG2 Receptors by Developing NK Cells Derived from Embryonic Stem Cells in Vitro1
    The Journal of Immunology Orderly and Nonstochastic Acquisition of CD94/NKG2 Receptors by Developing NK Cells Derived from Embryonic Stem Cells In Vitro1 Rebecca H. Lian,2* Motoi Maeda,2* Stefan Lohwasser,* Marc Delcommenne,† Toru Nakano,§ Russell E. Vance,¶ David H. Raulet,¶ and Fumio Takei3*‡ In mice there are two families of MHC class I-specific receptors, namely the Ly49 and CD94/NKG2 receptors. The latter receptors recognize the nonclassical MHC class I Qa-1b and are thought to be responsible for the recognition of missing-self and the maintenance of self-tolerance of fetal and neonatal NK cells that do not express Ly49. Currently, how NK cells acquire individual CD94/NKG2 receptors during their development is not known. In this study, we have established a multistep culture method to induce differentiation of embryonic stem (ES) cells into the NK cell lineage and examined the acquisition of CD94/NKG2 by NK cells as they differentiate from ES cells in vitro. ES-derived NK (ES-NK) cells express NK cell-associated proteins and they kill certain tumor cell lines as well as MHC class I-deficient lymphoblasts. They express CD94/NKG2 heterodimers, but not Ly49 molecules, and their cytotoxicity is inhibited by Qa-1b on target cells. Using RT-PCR analysis, we also report that the acquisition of these individual receptor gene expressions during different stages of differentiation from ES cells to NK cells follows a prede- termined order, with their order of acquisition being first CD94; subsequently NKG2D, NKG2A, and NKG2E; and finally, NKG2C. Single-cell RT-PCR showed coexpression of CD94 and NKG2 genes in most ES-NK cells, and flow cytometric analysis also detected CD94/NKG2 on most ES-NK cells, suggesting that the acquisition of these receptors by ES-NK cells in vitro is nonstochastic, orderly, and cumulative.
    [Show full text]
  • Cellular and Molecular Signatures in the Disease Tissue of Early
    Cellular and Molecular Signatures in the Disease Tissue of Early Rheumatoid Arthritis Stratify Clinical Response to csDMARD-Therapy and Predict Radiographic Progression Frances Humby1,* Myles Lewis1,* Nandhini Ramamoorthi2, Jason Hackney3, Michael Barnes1, Michele Bombardieri1, Francesca Setiadi2, Stephen Kelly1, Fabiola Bene1, Maria di Cicco1, Sudeh Riahi1, Vidalba Rocher-Ros1, Nora Ng1, Ilias Lazorou1, Rebecca E. Hands1, Desiree van der Heijde4, Robert Landewé5, Annette van der Helm-van Mil4, Alberto Cauli6, Iain B. McInnes7, Christopher D. Buckley8, Ernest Choy9, Peter Taylor10, Michael J. Townsend2 & Costantino Pitzalis1 1Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Departments of 2Biomarker Discovery OMNI, 3Bioinformatics and Computational Biology, Genentech Research and Early Development, South San Francisco, California 94080 USA 4Department of Rheumatology, Leiden University Medical Center, The Netherlands 5Department of Clinical Immunology & Rheumatology, Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands 6Rheumatology Unit, Department of Medical Sciences, Policlinico of the University of Cagliari, Cagliari, Italy 7Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK 8Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham B15 2WB, UK 9Institute of
    [Show full text]
  • The Journal of Experimental Medicine
    Published November 15, 2004 NKG2D Recognition and Perforin Effector Function Mediate Effective Cytokine Immunotherapy of Cancer Mark J. Smyth,1 Jeremy Swann,1 Janice M. Kelly,1 Erika Cretney,1 Wayne M. Yokoyama,2 Andreas Diefenbach,3 Thomas J. Sayers,4 and Yoshihiro Hayakawa1 1Cancer Immunology Program, Trescowthick Laboratories, Peter MacCallum Cancer Centre, East Melbourne, 8006 Victoria, Australia 2Howard Hughes Medical Institute, Washington University School of Medicine, St Louis, MO 63110 3Skirball Institute of Biomolecular Medicine, New York University Medical Center, New York, NY 10016 4Basic Research Program, SAIC-Frederick Inc., National Cancer Institute, Frederick, MD 21702 Abstract Downloaded from Single and combination cytokines offer promise in some patients with advanced cancer. Many spontaneous and experimental cancers naturally express ligands for the lectin-like type-2 trans- membrane stimulatory NKG2D immunoreceptor; however, the role this tumor recognition pathway plays in immunotherapy has not been explored to date. Here, we show that natural ex- pression of NKG2D ligands on tumors provides an effective target for some cytokine-stimulated NK cells to recognize and suppress tumor metastases. In particular, interleukin (IL)-2 or IL-12 jem.rupress.org suppressed tumor metastases largely via NKG2D ligand recognition and perforin-mediated cyto- toxicity. By contrast, IL-18 required tumor sensitivity to Fas ligand (FasL) and surprisingly did not depend on the NKG2D–NKG2D ligand pathway. A combination of IL-2 and IL-18 stimulated both perforin and FasL effector mechanisms with very potent effects. Cytokines that stimulated perforin-mediated cytotoxicity appeared relatively more effective against tumor metastases ex- pressing NKG2D ligands. These findings indicate that a rational choice of cytokines can be on March 23, 2015 made given the known sensitivity of tumor cells to perforin, FasL, and tumor necrosis factor– related apoptosis-inducing ligand and the NKG2D ligand status of tumor metastases.
    [Show full text]
  • Chimeric Non-Antigen Receptors in T Cell-Based Cancer Therapy
    Open access Review J Immunother Cancer: first published as 10.1136/jitc-2021-002628 on 3 August 2021. Downloaded from Chimeric non- antigen receptors in T cell- based cancer therapy Jitao Guo,1 Andrew Kent,1 Eduardo Davila1,2,3,4 To cite: Guo J, Kent A, Davila E. ABSTRACT including T cell trafficking, survival, prolifer- Chimeric non- antigen receptors Adoptively transferred T cell- based cancer therapies ation, differentiation, and effector functions, in T cell- based cancer therapy. have shown incredible promise in treatment of various would ideally fall under user-directed custom- Journal for ImmunoTherapy cancers. So far therapeutic strategies using T cells have of Cancer 2021;9:e002628. izable control. With these goals in mind, focused on manipulation of the antigen- recognition doi:10.1136/jitc-2021-002628 chimeric non- antigen receptors are being y itself, such as through selective expression machiner designed to provide supportive cosignaling of tumor- antigen specificT cell receptors or engineered Accepted 27 June 2021 antigen- recognition chimeric antigen receptors (CARs). for CAR or TCR antitumor T cell responses. While several CARs have been approved for treatment The domains, smaller motifs, and even key of hematopoietic malignancies, this kind of therapy has residues of natural immune receptors are been less successful in the treatment of solid tumors, in the essential functional subunits of each part due to lack of suitable tumor- specific targets, the receptor. Many domains and motifs exhibit immunosuppressive tumor microenvironment, and the high functional fidelity so long as their struc- inability of adoptively transferred cells to maintain their tural context is maintained, making them therapeutic potentials.
    [Show full text]
  • NKG2D Recognition and Perforin Effector Function Mediate Effective Cytokine Immunotherapy of Cancer Mark J
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2004 NKG2D recognition and perforin effector function mediate effective cytokine immunotherapy of cancer Mark J. Smyth Peter MacCallum Cancer Centre Jeremy Swann Peter MacCallum Cancer Centre Janice M. Kelly Peter MacCallum Cancer Centre Erika Cretney Peter MacCallum Cancer Centre Wayne M. Yokoyama Washington University School of Medicine in St. Louis See next page for additional authors Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Part of the Medicine and Health Sciences Commons Recommended Citation Smyth, Mark J.; Swann, Jeremy; Kelly, Janice M.; Cretney, Erika; Yokoyama, Wayne M.; Diefenbach, Andreas; Sayers, Thomas J.; and Hayakawa, Yoshihiro, ,"NKG2D recognition and perforin effector function mediate effective cytokine immunotherapy of cancer." Journal of Experimental Medicine.,. 1325-1335. (2004). https://digitalcommons.wustl.edu/open_access_pubs/613 This Open Access Publication is brought to you for free and open access by Digital Commons@Becker. It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact [email protected]. Authors Mark J. Smyth, Jeremy Swann, Janice M. Kelly, Erika Cretney, Wayne M. Yokoyama, Andreas Diefenbach, Thomas J. Sayers, and Yoshihiro Hayakawa This open access publication is available at Digital Commons@Becker: https://digitalcommons.wustl.edu/open_access_pubs/613 Published November
    [Show full text]
  • Diverse Functional Autoantibodies in Patients with COVID-19
    medRxiv preprint doi: https://doi.org/10.1101/2020.12.10.20247205; this version posted December 11, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY-NC-ND 4.0 International license . Diverse Functional Autoantibodies in Patients with COVID-19 Eric Y. Wang1,*, Tianyang Mao1,*, Jon Klein1,*, Yile Dai1,*, John D. Huck1, Feimei Liu1, Neil S. Zheng1, Ting Zhou1, Benjamin Israelow1, Patrick Wong1, Carolina Lucas1, Julio Silva1, Ji Eun Oh1, Eric Song1, Emily S. Perotti1, Suzanne Fischer1, Melissa Campbell5, John B. Fournier5, Anne L. Wyllie3, Chantal B. F. Vogels3, Isabel M. Ott3, Chaney C. Kalinich3, Mary E. Petrone3, Anne E. Watkins3, Yale IMPACT Team¶, Charles Dela Cruz4, Shelli F. Farhadian5, Wade L. Schulz6,7, Nathan D. Grubaugh3, Albert I. Ko3,5, Akiko Iwasaki1,3,8,#, Aaron M. Ring1,2,# 1 Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA 2 Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA 3 Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA 4 Department of Medicine, Section of Pulmonary and Critical Care Medicine, Yale School of Medicine, New Haven, CT, USA 5 Department of Internal Medicine (Infectious Diseases), Yale School of Medicine, New Haven, CT, USA 6 Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA 7 Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT, USA 8 Howard Hughes Medical Institute, Chevy Chase, MD, USA * These authors contributed equally to this work ¶ A list of authors and their affiliations appears at the end of the paper # Correspondence: [email protected] (A.M.R.); [email protected] (A.I.) 1 NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice.
    [Show full text]
  • Intratumoral Injection of SYNB1891, a Synthetic Biotic Medicine Designed
    Intratumoral injection of SYNB1891 A Synthetic Biotic medicine designed to activate the innate immune system. Therapy demonstrates target engagement in humans including intratumoral STING activation. Janku F, MD Anderson Cancer Center; Luke JJ, UPMC Hillman Cancer Center; Brennan AM, Synlogic; Riese RJ, Synlogic; Varterasian M, Pharmaceutical Consultant; Kuhn K, Synlogic; Sokolovska A, Synlogic; Strauss J, Mary Crowley Cancer Research Presented by Filip Janku, MD, PhD Study supported by Synlogic, Inc American Association for Cancer Research (AACR) April 2021 Introduction and Methods SYNB1891 Strain Phase 1 First-in-Human Clinical Trial • Live, modified strain of the probiotic E. coli • Enrolling patients with refractory advanced solid Nissle engineered to produce cyclic tumors or lymphoma dinucleotides (CDN) under hypoxia leading to stimulator of interferon genes (STING)- • Intratumoral (IT) injection of SYNB1891 on Days activation 1, 8 and 15 of the first 21-day cycle and then on Day 1 of each subsequent cycle. • Preferentially taken up by phagocytic antigen- presenting cells in tumors, activating • Dose escalation planned across 7 cohorts (1x106 complementary innate immune pathways – 1x109 live cells) with Arm 1 consisting of (direct CDN STING activation; cGAS-mediated SYNB1891 as monotherapy, and Arm 2 in STING activation and TLR4/MyD88 activation by combination with atezolizumab the bacterial chassis) SYNB1891 was safe and well-tolerated in heterogenous population Nov 2020: Interim Analysis IA Updated through 15 Mar 2021 15 Mar 2021:
    [Show full text]
  • Organ-Specific Features of Natural Killer Cells
    REVIEWS Organ-specific features of natural killer cells Fu-Dong Shi*‡, Hans-Gustaf Ljunggren§, Antonio La Cava|| and Luc Van Kaer¶ Abstract | Natural killer (NK) cells can be swiftly mobilized by danger signals and are among the earliest arrivals at target organs of disease. However, the role of NK cells in mounting inflammatory responses is often complex and sometimes paradoxical. Here, we examine the divergent phenotypic and functional features of NK cells, as deduced largely from experimental mouse models of pathophysiological responses in the liver, mucosal tissues, uterus, pancreas, joints and brain. Moreover, we discuss how organ-specific factors, the local microenvironment and unique cellular interactions may influence the organ-specific properties of NK cells. Infection and autoimmunity are two common patho- migration from the bone marrow through the blood to logical processes, during which inflammation might be the spleen, liver, lung and many other organs. The dis- elicited in an organ-specific manner. Microorganisms tribution of NK cells is not static because these cells can can infect specific organs and induce inflammatory recirculate between organs15. Owing to the expression of responses. Frequently, the host immune response against chemokine receptors, such as CC‑chemokine receptor 2 the invading pathogens may also cause tissue damage (CCR2), CCR5, CXC-chemokine receptor 3 (CXCR3) owing to bystander effects. Moreover, inflammation can and CX3C‑chemokine receptor 1 (CX3CR1), NK cells sometimes trigger organ-specific autoimmune diseases can respond to a large array of chemokines16, and thus or even malignant transformation of the affected organ. they can be recruited to distinct sites of inflammation17–25 *Department of Neurology, Among the many cell types of the immune system, and extravagate to the parenchyma or body cavities.
    [Show full text]
  • Spatial Transcriptomic Characterization of COVID-19 Pneumonitis Identifies Immune Pathways Related to Tissue Injury
    bioRxiv preprint doi: https://doi.org/10.1101/2021.06.21.449178; this version posted June 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Spatial transcriptomic characterization of COVID-19 pneumonitis identifies immune pathways related to tissue injury Cross AR1, de Andrea CE2, Landecho Acha MF2,3, Cerundolo L1, Etherington R4, Denney L4, Ho LP4, Roberts I5, Klenerman P3, Hester J1, Melero I2, Sansom SN6*, Issa F1* 1. Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK 2. Department of Immunology and Immunotherapy, Clínica de la Universdad de Navarra, Pamplona, Spain 3. Department of Internal Medicine, Clinica de la Universdad de Navarra, Pamplona, Spain 4. Nuffield Department of Medicine, University of Oxford, Oxford, UK 5. Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK 6. Kennedy Institute of Rheumatology, University of Oxford, UK *Joint senior authors. Corresponding author: [email protected] ABSTRACT Severe lung damage in COVID-19 is known to involve complex interactions between diverse populations of immune and stromal cells. In this study, we applied a spatial transcriptomics approach to better delineate the cells, pathways and genes responsible for promoting and perpetuating severe tissue pathology in COVID-19 pneumonitis. Guided by tissue histology and immunohistochemistry we performed a targeted sampling of dozens of regions representing a spectrum of diffuse alveolar damage from the post-mortem lung of three COVID-19 patients.
    [Show full text]
  • Prevalent Homozygous Deletions of Type I Interferon and Defensin Genes in Human Cancers Associate with Immunotherapy Resistance
    Author Manuscript Published OnlineFirst on April 4, 2018; DOI: 10.1158/1078-0432.CCR-17-3008 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Prevalent Homozygous Deletions of Type I Interferon and Defensin Genes in Human Cancers Associate with Immunotherapy Resistance Zhenqing Ye1; Haidong Dong2,3; Ying Li1; Tao Ma1,4; Haojie Huang4; Hon-Sing Leong4; Jeanette Eckel-Passow1; Jean-Pierre A. Kocher1; Han Liang5; LiguoWang1,4,* 1Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic, Rochester, Minnesota 55905, United States of America 2Department of Immunology, College of Medicine, Mayo Clinic, Rochester, Minnesota 55905, United States of America 3Department of Urology, Mayo Clinic, Rochester, Minnesota 55905, United States of America 4Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, United States of America 5Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States of America Running title: Pervasive Deletion of Interferon/Defensin in Human Cancers Keywords: Homozygous deletion, Type-I interferon, Defensin, immunotherapy resistance, cancer * Correspondence to: Liguo Wang, Ph.D. Associate Professor Division of Biomedical Statistics and Informatics, Mayo Clinic 200 1st St SW Rochester, MN 55905, USA Phone: +1-507-284-8728 Fax: +1-507-284-0745 Email: [email protected] The authors declare no potential conflicts of interest. 1 Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 4, 2018; DOI: 10.1158/1078-0432.CCR-17-3008 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
    [Show full text]