1
|
Zhou Y, Hubscher CH. Biomarker expression level changes within rectal gut-associated lymphoid tissues in spinal cord-injured rats. Immunohorizons 2025; 9:vlaf002. [PMID: 40048710 PMCID: PMC11884801 DOI: 10.1093/immhor/vlaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Neurogenic bowel dysfunction (NBD) is common after spinal cord injury (SCI). Gut-associated lymphoid tissue (GALT), an organized structure within the mucosal immune system, is important for the maintenance of gut homeostasis and body health and serves as the first line barrier/defense against diet antigens, commensal microbiota, pathogens, and toxins in mucosal areas. The current study examined gene expression levels along six segments of anorectal tissue using real-time polymerase chain reaction (RT-PCR) in uninjured rats (28-day sham surgical controls) and at both 28- and 42-days post-T9 contusion injury. Consistent with our previous report of functional regional differences in the ano-rectum, we demonstrate the existence of GALTs located primarily within the segment at 3-4.5 cm from the rectal dentate line (termed rectal GALTs-rGALTs) in shams with upregulated gene expression levels of multiple biomarkers, including B cell and T cell-related genes, major histocompatibility complex (MHC) class II molecules, and germinal center (GC)-related genes, which was further confirmed by histologic examination. In the same rectal tissue segment following T9 SCI, inflammation-related genes were upregulated at 28 days post-injury (DPI) indicating that microbial infection and inflammation of rGALTs modified structure and function of rGALTs, while at 42 DPI rGALTs exhibited resolution of inflammation and impaired structure/function for extrafollicular B cell responses. Taken together, our data suggest that rGALTs exists in rat rectum for homeostasis of gut microbiota/barrier. SCI induces microbial infection and inflammation in rectal tissues containing rGALTs, which could contribute to development of SCI-related gut microbiome dysbiosis, NBD, and systemic diseases.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Spinal Cord Injury Research Center, Louisville, KY, United States
| | - Charles H Hubscher
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Spinal Cord Injury Research Center, Louisville, KY, United States
| |
Collapse
|
2
|
Borowska MT, Liu LD, Caveney NA, Jude KM, Kim WJ, Masubuchi T, Hui E, Majzner RG, Garcia KC. Orientation-dependent CD45 inhibition with viral and engineered ligands. Sci Immunol 2024; 9:eadp0707. [PMID: 39454026 PMCID: PMC11537708 DOI: 10.1126/sciimmunol.adp0707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/01/2024] [Indexed: 10/27/2024]
Abstract
CD45 is a cell surface phosphatase that shapes the T cell receptor signaling threshold but does not have a known ligand. A family of adenovirus proteins, including E3/49K, exploits CD45 to evade immunity by binding to the extracellular domain of CD45, resulting in the suppression of T cell signaling. We determined the cryo-EM structure of this complex and found that the E3/49K protein is composed of three immunoglobulin domains assembled as "beads on a string" that compel CD45 into a closely abutted dimer by cross-linking the CD45 D3 domain, leading to steric inhibition of its intracellular phosphatase activity. Inspired by the E3/49K mechanism, we engineered CD45 surrogate ligands that can fine-tune T cell activation by dimerizing CD45 into different orientations and proximities. The adenovirus E3/49K protein has taught us that, despite a lack of a known ligand, CD45 activity can be modulated by extracellular dimerizing ligands that perturb its phosphatase activity and alter T cell responses.
Collapse
Affiliation(s)
- Marta T. Borowska
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94063, USA
| | - Liu D. Liu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94063, USA
| | - Nathanael A. Caveney
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94063, USA
| | - Kevin M. Jude
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94063, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94063, USA
| | - Won-Ju Kim
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Takeya Masubuchi
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Enfu Hui
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robbie G. Majzner
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - K. Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94063, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94063, USA
- Department of Structural Biology, Stanford University, Stanford, CA 94063, USA
| |
Collapse
|
3
|
Litvin U, Wang ECY, Stanton RJ, Fielding CA, Hughes J. Evolution of the Cytomegalovirus RL11 gene family in Old World monkeys and Great Apes. Virus Evol 2024; 10:veae066. [PMID: 39315401 PMCID: PMC11416908 DOI: 10.1093/ve/veae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/15/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
Cytomegalovirus (CMV) is a genus of herpesviruses, members of which share a long history of coevolution with their primate hosts including New World monkeys, Old World monkeys (OWMs), and Great Apes (GAs). These viruses are ubiquitous within their host populations and establish lifelong infection in most individuals. Although asymptomatic in healthy individuals, infection poses a significant risk to individuals with a weakened or underdeveloped immune system. The genome of human CMV is the largest among human-infecting viruses and comprises at least 15 separate gene families, which may have arisen by gene duplication. Within human CMV, the RL11 gene family is the largest. RL11 genes are nonessential in vitro but have immune evasion roles that are likely critical to persistence in vivo. These genes demonstrate an extreme level of inter-species and intra-strain sequence diversity, which makes it challenging to deduce the evolutionary relationships within this gene family. Understanding the evolutionary relationships of these genes, especially accurate ortholog identification, is essential for reconstructing ancestral genomes, deciphering gene repertoire and order, and enabling reliable functional analyses across the CMV species, thereby offering insights into evolutionary processes, genetic diversity, and the functional significance of genes. In this work, we combined in silico genome screening with sequence-based and structure-guided phylogenetic analysis to reconstruct the evolutionary history of the RL11 gene family. We confirmed that RL11 genes are unique to OWM and GA CMVs, showing that this gene family was formed by multiple early duplication events and later lineage-specific losses. We identified four main clades of RL11 genes and showed that their expansions were mainly lineage specific and happened independently in CMVs of GAs, African OWMs, and Asian OWMs. We also identified groups of orthologous genes across the CMV tree, showing that some human CMV-specific RL11 genes emerged before the divergence of human and chimpanzee CMVs but were subsequently lost in the latter. The extensive and dynamic species-specific evolution of this gene family suggests that their functions target elements of host immunity that have similarly coevolved during speciation.
Collapse
Affiliation(s)
- Ulad Litvin
- MRC-University of Glasgow Centre for Virus Research, Sir Michael Stoker Building, 464 Bearsden Road, Glasgow G61 1QH, United Kingdom
| | - Eddie C Y Wang
- Division of Infection and Immunity, Cardiff University School of Medicine, UHW Main Building, Heath Park, Cardiff CF14 4XN, United Kingdom
| | - Richard J Stanton
- Division of Infection and Immunity, Cardiff University School of Medicine, UHW Main Building, Heath Park, Cardiff CF14 4XN, United Kingdom
| | - Ceri A Fielding
- Division of Infection and Immunity, Cardiff University School of Medicine, UHW Main Building, Heath Park, Cardiff CF14 4XN, United Kingdom
| | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, Sir Michael Stoker Building, 464 Bearsden Road, Glasgow G61 1QH, United Kingdom
| |
Collapse
|
4
|
Hildenbrand A, Cramer P, Bertolotti M, Kaiser NS, Kläsener K, Nickel CM, Reth M, Heim A, Hengel H, Burgert HG, Ruzsics Z. Inhibition of B cell receptor signaling induced by the human adenovirus species D E3/49K protein. Front Immunol 2024; 15:1432226. [PMID: 39139562 PMCID: PMC11321000 DOI: 10.3389/fimmu.2024.1432226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction The early transcription unit 3 (E3) of human adenoviruses (HAdVs) encodes several immunoevasins, including the E3/49K protein, which is unique for species D of HAdVs. It is expressed as surface transmembrane protein and shed. E3/49K of HAdV-D64 binds to the protein tyrosine phosphatase surface receptor CD45, thereby modulating activation of T and NK cells. Methods Considering that E3/49K represents the most polymorphic viral protein among species D HAdVs, we demonstrate here that all tested E3/49K orthologs bind to the immunologically important regulator CD45. Thus, this feature is conserved regardless of the pathological associations of the respective HAdV types. Results It appeared that modulation of CD45 is a unique property restricted to HAdVs of species D. Moreover, E3/49K treatment inhibited B cell receptor (BCR) signaling and impaired BCR signal phenotypes. The latter were highly comparable to B cells having defects in the expression of CD45, suggesting E3/49K as a potential tool to investigate CD45 specific functions. Conclusion We identified B cells as new direct target of E3/49K-mediated immune modulation, representing a novel viral immunosubversive mechanism.
Collapse
Affiliation(s)
- Andreas Hildenbrand
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Precious Cramer
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Milena Bertolotti
- Signaling Research Centers CIBSS and BIOSS, University of Freiburg, Freiburg, Germany
- Navita S.r.l., University of Eastern Piedmont A. Avogadro, Novara, Italy
| | - Nathalie Sophia Kaiser
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kathrin Kläsener
- Department of Rheumatology and Clinical Immunology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Clara Muriel Nickel
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael Reth
- Signaling Research Centers CIBSS and BIOSS, University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Albert Heim
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Hartmut Hengel
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans-Gerhard Burgert
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Zsolt Ruzsics
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
5
|
Vlachava VM, Seirafian S, Fielding CA, Kollnberger S, Aicheler RJ, Hughes J, Baker A, Weekes MP, Forbes S, Wilkinson GWG, Wang ECY, Stanton RJ. HCMV-secreted glycoprotein gpUL4 inhibits TRAIL-mediated apoptosis and NK cell activation. Proc Natl Acad Sci U S A 2023; 120:e2309077120. [PMID: 38011551 PMCID: PMC10710050 DOI: 10.1073/pnas.2309077120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/07/2023] [Indexed: 11/29/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a paradigm of pathogen immune evasion and sustains lifelong persistent infection in the face of exceptionally powerful host immune responses through the concerted action of multiple immune-evasins. These reduce NK cell activation by inhibiting ligands for activating receptors, expressing ligands for inhibitory receptors, or inhibiting synapse formation. However, these functions only inhibit direct interactions with the infected cell. To determine whether the virus also expresses soluble factors that could modulate NK function at a distance, we systematically screened all 170 HCMV canonical protein-coding genes. This revealed that UL4 encodes a secreted and heavily glycosylated protein (gpUL4) that is expressed with late-phase kinetics and is capable of inhibiting NK cell degranulation. Analyses of gpUL4 binding partners by mass spectrometry identified an interaction with TRAIL. gpUL4 bound TRAIL with picomolar affinity and prevented TRAIL from binding its receptor, thus acting as a TRAIL decoy receptor. TRAIL is found in both soluble and membrane-bound forms, with expression of the membrane-bound form strongly up-regulated on NK cells in response to interferon. gpUL4 inhibited apoptosis induced by soluble TRAIL, while also binding to the NK cell surface in a TRAIL-dependent manner, where it blocked NK cell degranulation and cytokine secretion. gpUL4 therefore acts as an immune-evasin by inhibiting both soluble and membrane-bound TRAIL and is a viral-encoded TRAIL decoy receptor. Interestingly, gpUL4 could also suppress NK responses to heterologous viruses, suggesting that it may act as a systemic virally encoded immunosuppressive agent.
Collapse
Affiliation(s)
- Virginia-Maria Vlachava
- Infection and Immunity, School of Medicine, Cardiff University, CardiffCF14 4XN, United Kingdom
| | - Sepehr Seirafian
- Infection and Immunity, School of Medicine, Cardiff University, CardiffCF14 4XN, United Kingdom
| | - Ceri A. Fielding
- Infection and Immunity, School of Medicine, Cardiff University, CardiffCF14 4XN, United Kingdom
| | - Simon Kollnberger
- Infection and Immunity, School of Medicine, Cardiff University, CardiffCF14 4XN, United Kingdom
| | - Rebecca J. Aicheler
- Department of Biomedical Sciences, Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, CardiffCF5 2YB, United Kingdom
| | - Joseph Hughes
- Centre for Virus Research, School of Infection & Immunity, Glasgow University, GlasgowG61 1QH, United Kingdom
| | - Alexander Baker
- Infection and Immunity, School of Medicine, Cardiff University, CardiffCF14 4XN, United Kingdom
| | - Michael P. Weekes
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, CambridgeCB2 0XY, United Kingdom
| | - Simone Forbes
- Infection and Immunity, School of Medicine, Cardiff University, CardiffCF14 4XN, United Kingdom
| | - Gavin W. G. Wilkinson
- Infection and Immunity, School of Medicine, Cardiff University, CardiffCF14 4XN, United Kingdom
| | - Eddie C. Y. Wang
- Infection and Immunity, School of Medicine, Cardiff University, CardiffCF14 4XN, United Kingdom
| | - Richard J. Stanton
- Infection and Immunity, School of Medicine, Cardiff University, CardiffCF14 4XN, United Kingdom
| |
Collapse
|
6
|
Funk MA, Leitner J, Gerner MC, Hammerler JM, Salzer B, Lehner M, Battin C, Gumpelmair S, Stiasny K, Grabmeier-Pfistershammer K, Steinberger P. Interrogating ligand-receptor interactions using highly sensitive cellular biosensors. Nat Commun 2023; 14:7804. [PMID: 38016944 PMCID: PMC10684770 DOI: 10.1038/s41467-023-43589-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
Interactions of membrane-resident proteins are important targets for therapeutic interventions but most methods to study them are either costly, laborious or fail to reflect the physiologic interaction of membrane resident proteins in trans. Here we describe highly sensitive cellular biosensors as a tool to study receptor-ligand pairs. They consist of fluorescent reporter cells that express chimeric receptors harboring ectodomains of cell surface molecules and intracellular signaling domains. We show that a broad range of molecules can be integrated into this platform and we demonstrate its applicability to highly relevant research areas, including the characterization of immune checkpoints and the probing of cells for the presence of receptors or ligands. The platform is suitable to evaluate the interactions of viral proteins with host receptors and to test for neutralization capability of drugs or biological samples. Our results indicate that cellular biosensors have broad utility as a tool to study protein-interactions.
Collapse
Affiliation(s)
- Maximilian A Funk
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria
| | - Judith Leitner
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria.
| | - Marlene C Gerner
- Division of Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Jasmin M Hammerler
- Division of Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Benjamin Salzer
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- Christian Doppler Laboratory for Next Generation CAR T Cells, Vienna, Austria
| | - Manfred Lehner
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- Christian Doppler Laboratory for Next Generation CAR T Cells, Vienna, Austria
| | - Claire Battin
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria
| | - Simon Gumpelmair
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | - Peter Steinberger
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Pokoyski C, Baars W, Windheim M, Reubold TF, Zischke J, Brinkmann A, Kay-Fedorov PC, Schwinzer R. Expression of viral CD45 ligand E3/49K on porcine cells reduces human anti-pig immune responses. Sci Rep 2023; 13:17218. [PMID: 37821577 PMCID: PMC10567836 DOI: 10.1038/s41598-023-44316-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023] Open
Abstract
Transgenic expression of protective molecules in porcine cells and tissues is a promising approach to prevent xenograft rejection. Viruses have developed various strategies to escape the host's immune system. We generated porcine B cells (B cell line L23) expressing the human adenovirus protein E3/49K or the human cytomegalovirus protein pUL11 and investigated how human T, NK and B cell responses are affected by the expression of the viral proteins. Binding studies revealed that E3/49K and pUL11 interact with CD45 on human but not porcine peripheral blood mononuclear cells. T cell proliferation in response to L23-E3/49K cells was significantly reduced and accompanied by development of an anti-inflammatory cytokine milieu (low: TNF-alpha, IFN-gamma, IL-6; high: IL-4, IL-10). Human peripheral blood mononuclear cells which had been primed for four weeks by L23-E3/49K cells included an extended population of regulatory T cells. Cytotoxicity of effector T and natural killer cells against L23 cells was significantly reduced (40 to 50%) by E3/49K expression. B cell activation and antibody production to E3/49K expressing cells was also diminished. Surprisingly, pUL11 expression showed no effects. Reduction of human anti-pig immune responses by transgenic expression of selected viral genes may be a novel approach for protection of porcine xenografts.
Collapse
Affiliation(s)
- Claudia Pokoyski
- Surgical Research Laboratory, Hannover Medical School, Hannover, Germany.
| | - Wiebke Baars
- Surgical Research Laboratory, Hannover Medical School, Hannover, Germany
| | - Mark Windheim
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Thomas F Reubold
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Jasmin Zischke
- Institute of Virology, Hannover Medical School, and German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
| | - Antje Brinkmann
- Surgical Research Laboratory, Hannover Medical School, Hannover, Germany
| | - Penelope C Kay-Fedorov
- Institute of Virology, Hannover Medical School, and German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
| | - Reinhard Schwinzer
- Surgical Research Laboratory, Hannover Medical School, Hannover, Germany
| |
Collapse
|
8
|
Zeng J, Cao D, Yang S, Jaijyan DK, Liu X, Wu S, Cruz-Cosme R, Tang Q, Zhu H. Insights into the Transcriptome of Human Cytomegalovirus: A Comprehensive Review. Viruses 2023; 15:1703. [PMID: 37632045 PMCID: PMC10458407 DOI: 10.3390/v15081703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a widespread pathogen that poses significant risks to immunocompromised individuals. Its genome spans over 230 kbp and potentially encodes over 200 open-reading frames. The HCMV transcriptome consists of various types of RNAs, including messenger RNAs (mRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and microRNAs (miRNAs), with emerging insights into their biological functions. HCMV mRNAs are involved in crucial viral processes, such as viral replication, transcription, and translation regulation, as well as immune modulation and other effects on host cells. Additionally, four lncRNAs (RNA1.2, RNA2.7, RNA4.9, and RNA5.0) have been identified in HCMV, which play important roles in lytic replication like bypassing acute antiviral responses, promoting cell movement and viral spread, and maintaining HCMV latency. CircRNAs have gained attention for their important and diverse biological functions, including association with different diseases, acting as microRNA sponges, regulating parental gene expression, and serving as translation templates. Remarkably, HCMV encodes miRNAs which play critical roles in silencing human genes and other functions. This review gives an overview of human cytomegalovirus and current research on the HCMV transcriptome during lytic and latent infection.
Collapse
Affiliation(s)
- Janine Zeng
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA
| | - Di Cao
- Department of Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518052, China
| | - Shaomin Yang
- Department of Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518052, China
| | - Dabbu Kumar Jaijyan
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA
| | - Xiaolian Liu
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Songbin Wu
- Department of Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518052, China
| | - Ruth Cruz-Cosme
- Department of Microbiology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| | - Hua Zhu
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA
| |
Collapse
|
9
|
Ahmed MGT, Limmer A, Hartmann M. CD45RA and CD45RO Are Regulated in a Cell-Type Specific Manner in Inflammation and Sepsis. Cells 2023; 12:1873. [PMID: 37508538 PMCID: PMC10378241 DOI: 10.3390/cells12141873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
CD45 is a transmembrane glycoprotein that is located on the surface of all leukocytes and modulates both innate and adaptive immune system functions. In a recent study, inflammation modulated the CD45 expression in leukocytes, but the effect on the expression of CD45 subtypes is unknown. In the present study, we therefore investigated the effect of inflammatory conditions in humans (surgery, sepsis) and ex vivo incubation with lipopolysaccharides (LPS) on the expression of the subtypes CD45RA and CD45RO in granulocytes, lymphocytes, and monocytes. Whole blood samples were obtained from healthy volunteers, postoperative patients, and patients with sepsis at day 1 of diagnosis, respectively. Samples were incubated with fluorescent antibodies directed against CD45, CD45RA and CD45RO in the absence and presence of lipopolysaccharide and subjected to flow cytometry. In comparison to volunteers, CD45RA surface expression in postoperative and septic patients was reduced by 89% exclusively on granulocytes, but not on lymphocytes or monocytes. In contrast, CD45RO was exclusively reduced on lymphocytes, by 82%, but not on other cell types. Receiver operating characteristic curve analyses demonstrated that CD45RA (on granulocytes) and CD45RO (on lymphocytes) allow a good differentiation of volunteers and patients with sepsis (AUC = 0.9; p = 0.0001). The addition of LPS to the whole blood samples obtained from volunteers, postoperative patients, and septic patients markedly increased the CD45RO expression in granulocytes, lymphocytes, and monocytes. In contrast, LPS reduced CD45RA exclusively on monocytes. In conclusion, the surface expression of CD45RA and CD45RO is regulated in inflammation in a cell-type- and stimulus-specific manner. Considering that CD45 subtypes are critically involved in immune system signaling, the pathophysiologic and diagnostic implications warrant further investigation.
Collapse
Affiliation(s)
- Muhammad G T Ahmed
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- Anesthesia, Intensive Care and Pain Management, South Egypt Cancer Institute, Assiut University, Assiut 7111, Egypt
| | - Andreas Limmer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Matthias Hartmann
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
10
|
Windheim M, Reubold TF, Aichane K, Gaestel M, Burgert HG. Enforced Dimerization of CD45 by the Adenovirus E3/49K Protein Inhibits T Cell Receptor Signaling. J Virol 2023; 97:e0189822. [PMID: 37125921 PMCID: PMC10231199 DOI: 10.1128/jvi.01898-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/07/2023] [Indexed: 05/02/2023] Open
Abstract
Human adenoviruses (HAdVs) are widespread pathogens that generally cause mild infections in immunocompetent individuals but severe or even fatal diseases in immunocompromised patients. In order to counteract the host immune defenses, HAdVs encode various immunomodulatory proteins in the early transcription unit 3 (E3). The E3/49K protein is a highly glycosylated type I transmembrane protein uniquely expressed by species D HAdVs. Its N-terminal ectodomain sec49K is released by metalloprotease-mediated shedding at the cell surface and binds to the receptor-like protein tyrosine phosphatase CD45, a critical regulator of leukocyte activation and functions. It remained elusive which domains of CD45 and E3/49K are involved in the interaction and whether such an interaction can also occur on the cell surface with membrane-anchored full-length E3/49K. Here, we show that the two extracellular domains R1 and R2 of E3/49K bind to the same site in the domain d3 of CD45. This interaction enforces the dimerization of CD45, causing the inhibition of T cell receptor signaling. Intriguingly, the membrane-anchored E3/49K appears to be designed like a "molecular fishing rod" using an extended disordered region of E3/49K as a "fishing line" to bridge the distance between the plasma membrane of infected cells and the CD45 binding site on T cells to effectively position the domains R1 and R2 as baits for CD45 binding. This design strongly suggests that both secreted sec49K as well as membrane-anchored full-length E3/49K have immunomodulatory functions. The forced dimerization of CD45 may be applied as a therapeutic strategy in chronic inflammatory disorders and cancer. IMPORTANCE The battle between viruses and their hosts is an ongoing arms race. Whereas the host tries to detect and eliminate the virus, the latter counteracts such antiviral measures to replicate and spread. Adenoviruses have evolved various mechanisms to evade the human immune response. The E3/49K protein of species D adenoviruses mediates the inhibition of immune cell function via binding to the protein tyrosine phosphatase CD45. Here, we show that E3/49K triggers the dimerization of CD45 and thereby inhibits its phosphatase activity. Intriguingly, the membrane-anchored E3/49K seems to be designed like a "molecular fishing rod" with the two CD45 binding domains of E3/49K as baits positioned at the end of an extended disordered region reminiscent of a fishing line. The adenoviral strategy to inhibit CD45 activity by forced dimerization may be used for therapeutic intervention in autoimmune diseases or to prevent graft rejection after transplantation.
Collapse
Affiliation(s)
- Mark Windheim
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Thomas F. Reubold
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Khadija Aichane
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Hans-Gerhard Burgert
- Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| |
Collapse
|
11
|
Human Cytomegalovirus pUL11, a CD45 Ligand, Disrupts CD4 T Cell Control of Viral Spread in Epithelial Cells. mBio 2022; 13:e0294622. [PMID: 36445084 PMCID: PMC9765415 DOI: 10.1128/mbio.02946-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Human cytomegalovirus (HCMV) encodes numerous immunomodulatory genes that facilitate its persistence. Previously described mechanisms by which HCMV avoids T cell control typically involve evasion of detection by infected cells. Here, we show that the virus also inhibits T cells directly via an interaction between the pUL11 glycoprotein on infected cells and the CD45 phosphatase on T cells. The antiviral functions of CD4 T cells are impaired as a result of this interaction, largely via induced interleukin 10 (IL-10) secretion in the CD4 T cell central memory compartment, resulting in enhanced viral spread. This establishes CD45 as an inhibitory receptor that regulates antiviral T cell functions and has parallels with the manipulation of natural killer (NK) cells by HCMV. By coculturing donor T cells with HCMV-infected epithelial cells, we observed that CD4 T cells can respond to epithelial cell antigen presentation and can control HCMV spread via cytolytic and cytokine-dependent mechanisms. pUL11 impairs both mechanisms. We showed that pUL11-induced IL-10 secretion requires IL-2, mTOR, and T cell receptor signaling. This characterization of the effects of the pUL11-CD45 interaction may allow for the development of new antiviral therapies and treatments for inflammatory disorders. IMPORTANCE Human cytomegalovirus (HCMV) is adept at avoiding its host's immune defenses, both by evading detection and by directly inhibiting immune cells. This can lead to a loss of control of the infection, and dangerous disease can result, particularly in cases in which an individual's immune system is immature, weak, or suppressed. T cells form a crucial part of the response to HCMV and are used in cellular HCMV therapies. We show that an interaction between a viral glycoprotein (pUL11) and a T cell surface receptor (CD45) impairs T cell memory functions and allows for increased viral spread. This defines a new immunomodulatory strategy for the virus as well as a new T cell regulatory mechanism. These results are important, as they increase our understanding of how T cells function and how HCMV disrupts them. This will allow for the development of new antiviral therapies that restore T cell functions and indicates a new target for controlling pathological T cell disorders.
Collapse
|
12
|
Rousselière A, Gérard N, Delbos L, Guérif P, Giral M, Bressollette-Bodin C, Charreau B. Distinctive phenotype for HLA-E- versus HLA-A2-restricted memory CD8 αβT cells in the course of HCMV infection discloses features shared with NKG2C +CD57 +NK and δ2 -γδT cell subsets. Front Immunol 2022; 13:1063690. [PMID: 36532017 PMCID: PMC9752567 DOI: 10.3389/fimmu.2022.1063690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/10/2022] [Indexed: 12/04/2022] Open
Abstract
The human cytomegalovirus (HCMV) triggers both innate and adaptive immune responses, including protective CD8+ αβT cells (CD8T) that contributes to the control of the infection. In addition to CD8T restricted by classical HLA class Ia molecules, HCMV also triggers CD8T recognizing peptides from the HCMV UL40 leader peptide and restricted by HLA-E molecules (HLA-EUL40 CD8T). This study investigated the frequency, phenotype and functions of HLA-EUL40 CD8T in comparison to the immunodominant HLA-A2pp65 CD8T upon acute (primary or secondary infection) or chronic infection in kidney transplant recipients (KTR) and in seropositive (HCMV+) healthy volunteer (HV) hosts. The frequency of hosts with detected HLA-EUL40 CD8T was similar after a primary infection (24%) and during viral latency in HCMV+ HV (26%) and equal to the frequency of HLA-A2pp65 CD8T cells in both conditions (29%). Both CD8T subsets vary from 0.1% to >30% of total circulating CD8T according to the host. Both HLA-EUL40 and HLA-A2pp65 CD8T display a phenotype specific of CD8+ TEMRA (CD45RA+/CCR7-) but HLA-EUL40 CD8T express distinctive level for CD3, CD8 and CD45RA. Tim3, Lag-3, 4-1BB, and to a lesser extend 2B4 are hallmarks for T cell priming post-primary infection while KLRG1 and Tigit are markers for restimulated and long lived HCMV-specific CD8T responses. These cell markers are equally expressed on HLA-EUL40 and HLA-A2pp65 CD8T. In contrast, CD56 and PD-1 are cell markers discriminating memory HLA-E- from HLA-A2-restricted CD8T. Long lived HLA-EUL40 display higher proliferation rate compared to HLA-A2pp65 CD8T consistent with elevated CD57 expression. Finally, a comparative immunoprofiling indicated that HLA-EUL40 CD8T, divergent from HLA-A2pp65 CD8T, share the expression of CD56, CD57, NKG2C, CD158 and the lack of PD-1 with NKG2C+CD57+ NK and δ2-γδT cells induced in response to HCMV and thus defines a common immunopattern for these subsets.
Collapse
Affiliation(s)
- Amélie Rousselière
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, UMR 1064, Nantes, France
| | - Nathalie Gérard
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, UMR 1064, Nantes, France
| | - Laurence Delbos
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, UMR 1064, Nantes, France
| | - Pierrick Guérif
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, France
| | - Magali Giral
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, UMR 1064, Nantes, France,CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, France
| | - Céline Bressollette-Bodin
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, UMR 1064, Nantes, France,CHU Nantes, Nantes Université, Laboratoire de Virologie, Nantes, France
| | - Béatrice Charreau
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, UMR 1064, Nantes, France,CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, France,*Correspondence: Béatrice Charreau,
| |
Collapse
|
13
|
Differential Regulation of CD45 Expression on Granulocytes, Lymphocytes, and Monocytes in COVID-19. J Clin Med 2022; 11:jcm11144219. [PMID: 35887979 PMCID: PMC9318847 DOI: 10.3390/jcm11144219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 02/01/2023] Open
Abstract
CD45 is a transmembrane glycoprotein and protein tyrosine phosphatase expressed on the surface of all nucleated hematopoietic cells. While there is increasing evidence demonstrating the involvement of CD45 in immune system regulation, no information on CD45 expression in inflammation and sepsis is currently available. Therefore, we determined the CD45 surface expression on granulocytes, lymphocytes, and monocytes in patients with COVID-19 and healthy volunteers in both absence and presence of lipopolysaccharide (LPS). Following approval by the local ethics committee, whole blood samples were obtained from patients with COVID-19 infection on day 1 of hospital admission and healthy volunteers. Samples were incubated in absence and presence of LPS and CD45 was measured in granulocytes, lymphocytes, and monocytes using flow cytometry. In comparison with healthy individuals, COVID-19 patients showed an increased CD45 expression on the surface of granulocytes (+35%, p < 0.02) and lymphocytes (+39%, p < 0.0001), but a reduced CD45 expression on monocytes (−35%, p < 0.0001). LPS incubation of whole blood from healthy individuals increased the CD45 expression on granulocytes (+430%, p < 0.0001), lymphocytes (+32%, p = 0.0012), and monocytes (+36%, p = 0.0005), respectively. LPS incubation of whole blood samples from COVID-19 patients increased the CD45 expression on granulocytes and monocytes, and decreased the CD45 expression on lymphocytes. In conclusion, CD45 expression on leucocytes is altered: (1) in COVID-19 patients, and (2) in in vitro endotoxemia in a complex cell-specific way, thus representing a new immunoregulatory mechanism.
Collapse
|
14
|
Alon D, Paitan Y, Robinson E, Ganor N, Lipovetsky J, Yerushalmi R, Cohen CJ, Raiter A. Downregulation of CD45 Signaling in COVID-19 Patients Is Reversed by C24D, a Novel CD45 Targeting Peptide. Front Med (Lausanne) 2021; 8:675963. [PMID: 34414199 PMCID: PMC8369232 DOI: 10.3389/fmed.2021.675963] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/09/2021] [Indexed: 02/05/2023] Open
Abstract
CD45, the predominant transmembrane tyrosine phosphatase in leukocytes, is required for the efficient induction of T cell receptor signaling and activation. We recently reported that the CD45-intracellular signals in peripheral blood mononuclear cells (PBMCs) of triple negative breast cancer (TNBC) patients are inhibited. We also reported that C24D, an immune modulating therapeutic peptide, binds to CD45 on immune-suppressed cells and resets the functionality of the immune system via the CD45 signaling pathway. Various studies have demonstrated that also viruses can interfere with the functions of CD45 and that patients with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) are immune-suppressed. Given the similarity between the role of CD45 in viral immune suppression and our findings on TNBC, we hypothesized that the C24D peptide may have a similar "immune-resetting" effect on PBMCs from COVID-19 patients as it did on PBMCs from TNBC patients. We tested this hypothesis by comparing the CD45/TCR intracellular signaling in PBMCs from ten COVID-19 patients vs. PBMCs from ten healthy volunteers. Herein, we report our findings, demonstrating the immune reactivating effect of C24D via the phosphorylation of the tyrosine 505 and 394 in Lck, the tyrosine 493 in ZAP-70 and the tyrosine 172 in VAV-1 proteins in the CD45 signaling pathway. Despite the relatively small number of patients in this report, the results demonstrate that C24D rescued CD45 signaling. Given the central role played by CD45 in the immune system, we suggest CD45 as a potential therapeutic target.
Collapse
Affiliation(s)
- Danny Alon
- Department of Medicine A, Meir Medical Center, Kfar Saba, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yossi Paitan
- Microbiology Laboratory, Meir Medical Center, Kfar Saba, Israel
| | - Eyal Robinson
- Department of Medicine B, Meir Medical Center, Kfar Saba, Israel
| | - Nirit Ganor
- Microbiology Laboratory, Meir Medical Center, Kfar Saba, Israel
| | - Julia Lipovetsky
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Petach Tikva, Israel
| | - Rinat Yerushalmi
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Petach Tikva, Israel
| | - Cyrille J. Cohen
- Laboratory of Tumor Immunotherapy, The Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan, Israel
| | - Annat Raiter
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Petach Tikva, Israel
| |
Collapse
|
15
|
Abstract
Like all herpesviruses, the roseoloviruses (HHV6A, -6B, and -7) establish lifelong infection within their host, requiring these viruses to evade host antiviral responses. One common host-evasion strategy is the downregulation of host-encoded, surface-expressed glycoproteins. Roseoloviruses have been shown to evade the host immune response by downregulating NK-activating ligands, class I MHC, and the TCR/CD3 complex. To more globally identify glycoproteins that are differentially expressed on the surface of HHV6A-infected cells, we performed cell surface capture of N-linked glycoproteins present on the surface of T cells infected with HHV6A, and compared these to proteins present on the surface of uninfected T cells. We found that the protein tyrosine phosphatase CD45 is downregulated in T cells infected with HHV6A. We also demonstrated that CD45 is similarly downregulated in cells infected with HHV7. CD45 is essential for signaling through the T cell receptor and, as such, is necessary for developing a fully functional immune response. Interestingly, the closely related betaherpesviruses human cytomegalovirus (HCMV) and murine cytomegalovirus (MCMV) have also separately evolved unique mechanisms to target CD45. While HCMV and MCMV target CD45 signaling and trafficking, HHV6A acts to downregulate CD45 transcripts. IMPORTANCE Human herpesviruses-6 and -7 infect essentially 100% of the world's population before the age of 5 and then remain latent or persistent in their host throughout life. As such, these viruses are among the most pervasive and stealthy of all viruses. Host immune cells rely on the presence of surface-expressed proteins to identify and target virus-infected cells. Here, we investigated the changes that occur to proteins expressed on the cell surface of T cells after infection with human herpesvirus-6A. We discovered that HHV-6A infection results in a reduction of CD45 on the surface of infected T cells and impaired activation in response to T cell receptor stimulation.
Collapse
|
16
|
Al Barashdi MA, Ali A, McMullin MF, Mills K. Protein tyrosine phosphatase receptor type C (PTPRC or CD45). J Clin Pathol 2021; 74:548-552. [PMID: 34039664 PMCID: PMC8380896 DOI: 10.1136/jclinpath-2020-206927] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 02/16/2021] [Indexed: 12/21/2022]
Abstract
The leucocyte common antigen, protein tyrosine phosphatase receptor type C (PTPRC), also known as CD45, is a transmembrane glycoprotein, expressed on almost all haematopoietic cells except for mature erythrocytes, and is an essential regulator of T and B cell antigen receptor-mediated activation. Disruption of the equilibrium between protein tyrosine kinase and phosphatase activity (from CD45 and others) can result in immunodeficiency, autoimmunity, or malignancy. CD45 is normally present on the cell surface, therefore it works upstream of a large signalling network which differs between cell types, and thus the effects of CD45 on these cells are also different. However, it is becoming clear that CD45 plays an essential role in the innate immune system and this is likely to be a key area for future research. In this review of PTPRC (CD45), its structure and biological activities as well as abnormal expression of CD45 in leukaemia and lymphoma will be discussed.
Collapse
Affiliation(s)
| | - Ahlam Ali
- Patrick G Johnston Centre for Cancer Research (PGJCCR), Queen's University Belfast, Belfast, UK
| | | | - Ken Mills
- Patrick G Johnston Centre for Cancer Research (PGJCCR), Queen's University Belfast, Belfast, UK
| |
Collapse
|
17
|
Li P, He F, Wu C, Zhao G, Hardwidge PR, Li N, Peng Y. Transcriptomic Analysis of Chicken Lungs Infected With Avian and Bovine Pasteurella multocida Serotype A. Front Vet Sci 2020; 7:452. [PMID: 32851030 PMCID: PMC7433353 DOI: 10.3389/fvets.2020.00452] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022] Open
Abstract
Pasteurella multocida (P. multocida) is a common animal pathogen responsible for many animal diseases. Strains from different hosts exhibit disparate degrees of effect in other species. Here, we characterize an avian P. multocida serogroup A strain (PmQ) showing high lethality to chickens and a bovine P. multocida serogroup A strain (PmCQ2) with no lethality to chickens. We used RNA-seq to profile the transcriptomes of chicken lungs infected with PmQ and PmCQ2. A total of 1,649 differentially expressed genes (DEGs) due to PmQ infection (831 upregulated genes and 818 downregulated genes) and 1427 DEGs (633 upregulated genes and 794 downregulated genes) due to PmCQ2 infection were identified. Functional analysis of these DEGs demonstrated that the TNF signaling pathway, the toll-like receptor signaling pathway, complement and coagulation cascades, and cytokine–cytokine receptor interaction were both enriched in PmQ and PmCQ2 infection. STAT and apoptosis signaling pathways were uniquely enriched by PmQ infection, and the NOD-like receptor signaling pathway was enriched only by PmCQ2 infection. Cell-type enrichment analysis of the transcriptomes showed that immune cells, including macrophages and granulocytes, were enriched in both infection groups. Collectively, our study profiled the transcriptomic response of chicken lungs infected with P. multocida and provided valuable information to understand the chicken responses to P. multocida infection.
Collapse
Affiliation(s)
- Pan Li
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Fang He
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Chenlu Wu
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Guangfu Zhao
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science and Technology, Southwest University, Chongqing, China.,The College of Life Sciences, Sichuan University, Chengdu, China
| | - Philip R Hardwidge
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Nengzhang Li
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yuanyi Peng
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science and Technology, Southwest University, Chongqing, China
| |
Collapse
|
18
|
Poole E, Neves TC, Oliveira MT, Sinclair J, da Silva MCC. Human Cytomegalovirus Interleukin 10 Homologs: Facing the Immune System. Front Cell Infect Microbiol 2020; 10:245. [PMID: 32582563 PMCID: PMC7296156 DOI: 10.3389/fcimb.2020.00245] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Human Cytomegalovirus (HCMV) can cause a variety of health disorders that can lead to death in immunocompromised individuals and neonates. The HCMV lifecycle comprises both a lytic (productive) and a latent (non-productive) phase. HCMV lytic infection occurs in a wide range of terminally differentiated cell types. HCMV latency has been less well-studied, but one characterized site of latency is in precursor cells of the myeloid lineage. All known viral genes are expressed during a lytic infection and a subset of these are also transcribed during latency. The UL111A gene which encodes the viral IL-10, a homolog of the human IL-10, is one of these genes. During infection, different transcript isoforms of UL111A are generated by alternative splicing. The most studied of the UL111A isoforms are cmvIL-10 (also termed the "A" transcript) and LAcmvIL-10 (also termed the "B" transcript), the latter being a well-characterized latency associated transcript. Both isoforms can downregulate MHC class II, however they differ in a number of other immunomodulatory properties, such as the ability to bind the IL10 receptor and induce signaling through STAT3. There are also a number of other isoforms which have been identified which are expressed by differential splicing during lytic infection termed C, D, E, F, and G, although these have been less extensively studied. HCMV uses the viral IL-10 proteins to manipulate the immune system during lytic and latent phases of infection. In this review, we will discuss the literature on the viral IL-10 transcripts identified to date, their encoded proteins and the structures of these proteins as well as the functional properties of all the different isoforms of viral IL-10.
Collapse
Affiliation(s)
- Emma Poole
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tainan Cerqueira Neves
- Center for Natural and Humanities Sciences, Federal University of ABC (UFABC), São Bernardo do Campo, Brazil
| | - Martha Trindade Oliveira
- Center for Natural and Humanities Sciences, Federal University of ABC (UFABC), São Bernardo do Campo, Brazil
| | - John Sinclair
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
19
|
Abstract
: The use of cytomegalovirus (CMV) as a vaccine vector to express antigens against multiple infectious diseases, including simian immunodeficiency virus, Ebola virus, plasmodium, and mycobacterium tuberculosis, in rhesus macaques has generated extraordinary levels of protective immunity against subsequent pathogenic challenge. Moreover, the mechanisms of immune protection have altered paradigms about viral vector-mediated immunity against ectopically expressed vaccine antigens. Further optimization of CMV-vectored vaccines, particularly as this approach moves to human clinical trials will be augmented by a more complete understanding of how CMV engenders mechanisms of immune protection. This review summarizes the particulars of the specific CMV vaccine vector that has been used to date (rhesus CMV strain 68-1) in relation to CMV natural history.
Collapse
|
20
|
Characterization of pUL5, an HCMV protein interacting with the cellular protein IQGAP1. Virology 2019; 540:57-65. [PMID: 31739185 DOI: 10.1016/j.virol.2019.10.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 01/01/2023]
Abstract
Among the Herpesviridae, human cytomegalovirus (HCMV) owns the largest genome and displays a huge coding potential. Here, we characterized the UL5 gene product (pUL5) of the clinical isolate TR strain. The protein was predicted as a 166-amino-acid membrane protein with a theoretical mass of 19 kDa. Recombinant virus expressing pUL5 with a tag allowed the identification of two pUL5 non-glycosylated species of approximately 19 and 9 kDa, expressed with early and late kinetic respectively. Experiments in infection confirmed that the lower molecular weight species was translated from an internal ATG in the UL5 open reading frame. Confocal microscopy analysis showed that pUL5 localized within the assembly compartment, but is not incorporated in the virion, as shown by Western blot on purified viral particles. Finally, pull-down experiments coupled with mass spectrometry analysis identified IQGAP1 as a pUL5 interactor, giving new hints on possible roles of pUL5 during HCMV infection.
Collapse
|
21
|
Kolb P, Sijmons S, McArdle MR, Taher H, Womack J, Hughes C, Ventura A, Jarvis MA, Stahl-Hennig C, Hansen S, Picker LJ, Malouli D, Hengel H, Früh K. Identification and Functional Characterization of a Novel Fc Gamma-Binding Glycoprotein in Rhesus Cytomegalovirus. J Virol 2019; 93:e02077-18. [PMID: 30487278 PMCID: PMC6364020 DOI: 10.1128/jvi.02077-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 12/20/2022] Open
Abstract
Receptors recognizing the Fc part of immunoglobulin G (FcγRs) are key determinants in antibody-mediated immune responses. Members of the Herpesviridae interfere with this immune regulatory network by expressing viral FcγRs (vFcγRs). Human cytomegalovirus (HCMV) encodes four distinct vFcγRs that differ with respect to their IgG subtype specificity and their impact on antibody-mediated immune function in vitro The impact of vFcγRs on HCMV pathogenesis and immunomodulation in vivo is not known. The closest evolutionary animal model of HCMV is rhesus CMV (RhCMV) infection of rhesus macaques. To enable the characterization of vFcγR function in this model, we studied IgG binding by RhCMV. We show that lysates of RhCMV-infected cells contain an IgG-binding protein of 30 kDa encoded by the gene Rh05 that is a predicted type I glycoprotein belonging to the RL11 gene family. Upon deletion of Rh05, IgG-Fc binding by RhCMV strain 68-1 is lost, whereas ectopic expression of Rh05 results in IgG binding to transfected cells consistent with Rh05 being a vFcγR. Using a set of reporter cell lines stably expressing human and rhesus FcγRs, we further demonstrate that Rh05 antagonizes host FcγR activation. Compared to Rh05-intact RhCMV, RhCMVΔRh05 showed an increased activation of host FcγR upon exposure of infected cells to IgG from RhCMV-seropositive animals, suggesting that Rh05 protects infected cells from opsonization and IgG-dependent activation of host FcγRs. However, antagonizing host FcγR activation by Rh05 was not required for the establishment and maintenance of infection of RhCMV, even in a seropositive host, as shown by the induction of T cell responses to heterologous antigens expressed by RhCMV lacking the gene region encoding Rh05. In contrast to viral evasion of natural killer cells or T cell recognition, the evasion of antibody-mediated effects does not seem to be absolutely required for infection or reinfection. The identification of the first vFcγR that efficiently antagonizes host FcγR activation in the RhCMV genome will thus permit more detailed studies of this immunomodulatory mechanism in promoting viral dissemination in the presence of natural or vaccine-induced humoral immunity.IMPORTANCE Rhesus cytomegalovirus (RhCMV) offers a unique model for studying human cytomegalovirus (HCMV) pathogenesis and vaccine development. RhCMV infection of nonhuman primates greatly broadened the understanding of mechanisms by which CMVs evade or reprogram T cell and natural killer cell responses in vivo However, the role of humoral immunity and viral modulation of anti-CMV antibodies has not been studied in this model. There is evidence from in vitro studies that HCMVs can evade humoral immunity. By gene mapping and with the help of a novel cell-based reporter assay system we characterized the first RhCMV encoded IgG-Fcγ binding glycoprotein as a potent antagonist of rhesus FcγR activation. We further demonstrate that, unlike evasion of T cell immunity, this viral Fcγ receptor is not required to overcome anti-CMV immunity to establish secondary infections. These findings enable more detailed studies of the in vivo consequences of CMV evasion from IgG responses in nonhuman primate models.
Collapse
Affiliation(s)
- Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Steven Sijmons
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Matthew R McArdle
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Husam Taher
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Jennie Womack
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Colette Hughes
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Abigail Ventura
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Michael A Jarvis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | - Scott Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| |
Collapse
|
22
|
Human Cytomegalovirus Encodes a Novel FLT3 Receptor Ligand Necessary for Hematopoietic Cell Differentiation and Viral Reactivation. mBio 2018; 9:mBio.00682-18. [PMID: 29691342 PMCID: PMC5915732 DOI: 10.1128/mbio.00682-18] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The ability of human cytomegalovirus (HCMV) to reactivate from latent infection of hematopoietic progenitor cells (HPCs) is intimately linked to cellular differentiation. HCMV encodes UL7 that our group has shown is secreted from infected cells and induces angiogenesis. In this study, we show that UL7 is a ligand for Fms-like tyrosine kinase 3 receptor (Flt-3R), a well-known critical factor in HPC differentiation. We observed that UL7 directly binds Flt-3R and induces downstream signaling cascades, including phosphatidylinositol 3-kinase (PI3K)/Akt and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathways. Importantly, we show that UL7 protein induces differentiation of both CD34+ HPCs and CD14+ monocytes. Last, we show that an HCMV mutant lacking UL7 fails to reactivate in CD34+ HPCs in vitro as well as in humanized mice. These observations define the first virally encoded differentiation factor with significant implications not only for HCMV reactivation but also for alteration of the hematopoietic compartment in transplant patients.IMPORTANCE Human cytomegalovirus (HCMV) remains a significant cause of morbidity and mortality in allogeneic hematopoietic stem cell transplant recipients. CD34+ hematopoietic progenitor cells (HPCs) represent a critical reservoir of latent HCMV in the transplant population, thereby providing a source of virus for dissemination to visceral organs. HCMV reactivation has been linked to HPC/myeloid cellular differentiation; however, the mechanisms involved in these events are poorly understood at the molecular level. In this study, we show that a viral protein is a ligand for Fms-like tyrosine kinase 3 receptor (Flt-3R) and that the binding of HCMV UL7 to the Flt-3R triggers HPC and monocyte differentiation. Moreover, the loss of UL7 prevents viral reactivation in HPCs in vitro as well as in humanized mice. These observations define the first virally encoded differentiation factor with significant implications not only for HCMV reactivation but also for alteration of the hematopoietic compartment in transplant patients.
Collapse
|
23
|
A Prominent Role of the Human Cytomegalovirus UL8 Glycoprotein in Restraining Proinflammatory Cytokine Production by Myeloid Cells at Late Times during Infection. J Virol 2018; 92:JVI.02229-17. [PMID: 29467314 DOI: 10.1128/jvi.02229-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/14/2018] [Indexed: 01/21/2023] Open
Abstract
Human cytomegalovirus (HCMV) persistence in infected individuals relies on a plethora of mechanisms to efficiently reduce host immune responses. To that end, HCMV uses a variety of gene products, some of which have not been identified yet. Here we characterized the UL8 gene, which consists of two exons, sharing the first with the HCMV RL11 family member UL7 UL8 is a transmembrane protein with an N-terminal immunoglobulin (Ig)-like domain in common with UL7 but with an extended stalk and a distinctive cytoplasmic tail. The UL8 open reading frame gives rise to a heavily glycosylated protein predominantly expressed on the cell surface, from where it can be partially endocytosed and subsequently degraded. Infections with UL8-tagged viruses indicated that UL8 was synthesized with late-phase kinetics. By virtue of its highly conserved Ig-like domain, this viral protein interacted with a surface molecule present on activated neutrophils. Notably, when ectopically expressed in THP-1 myeloid cells, UL8 was able to significantly reduce the production of a variety of proinflammatory cytokines. Mutations in UL8 indicated that this functional effect was mediated by the cell surface expression of its Ig-like domain. To investigate the impact of the viral protein in the infection context, we engineered HCMVs lacking the UL8 gene and demonstrated that UL8 decreases the release of a large number of proinflammatory factors at late times after infection of THP-1 cells. Our data indicate that UL8 may exert an immunosuppressive role key for HCMV survival in the host.IMPORTANCE HCMV is a major pathogen that causes life-threatening diseases and disabilities in infected newborns and immunocompromised individuals. Containing one of the largest genomes among all reported human viruses, HCMV encodes an impressive repertoire of gene products. However, the functions of a large proportion of them still remain unknown, a fact that complicates the design of new therapeutic approaches to prevent or treat HCMV-associated diseases. In this report, we have conducted an extensive study of UL8, one of the previously uncharacterized HCMV open reading frames. We found that the UL8 protein is expressed at late times postinfection and utilized by HCMV to reduce the production of proinflammatory factors by infected myeloid cells. Thus, the work presented here points to a key role of UL8 as a novel HCMV immune modulator capable of restraining host antiviral defenses.
Collapse
|
24
|
Cytomegalovirus induces HLA-class-II-restricted alloreactivity in an acute myeloid leukemia cell line. PLoS One 2018; 13:e0191482. [PMID: 29377903 PMCID: PMC5788343 DOI: 10.1371/journal.pone.0191482] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 01/06/2018] [Indexed: 02/07/2023] Open
Abstract
Cytomegalovirus (HCMV) reactivation is found frequently after allogeneic hematopoietic stem cell transplantation (alloSCT) and is associated with an increased treatment-related mortality. Recent reports suggest a link between HCMV and a reduced risk of cancer progression in patients with acute leukemia or lymphoma after alloSCT. Here we show that HCMV can inhibit the proliferation of the acute myeloid leukemia cell line Kasumi-1 and the promyeloid leukemia cell line NB4. HCMV induced a significant up-regulation of HLA-class-II-molecules, especially HLA-DR expression and an increase of apoptosis, granzyme B, perforin and IFN-γ secretion in Kasumi-1 cells cocultured with peripheral blood mononuclear cells (PBMCs). Indolamin-2,3-dioxygenase on the other hand led only to a significant dose-dependent effect on IFN-γ secretion without effects on proliferation. The addition of CpG-rich oligonucleotides and ganciclovir reversed those antiproliferative effects. We conclude that HCMV can enhance alloreactivity of PBMCs against Kasumi-1 and NB4 cells in vitro. To determine if this phenomenon may be clinically relevant further investigations will be required.
Collapse
|
25
|
Zischke J, Mamareli P, Pokoyski C, Gabaev I, Buyny S, Jacobs R, Falk CS, Lochner M, Sparwasser T, Schulz TF, Kay-Fedorov PC. The human cytomegalovirus glycoprotein pUL11 acts via CD45 to induce T cell IL-10 secretion. PLoS Pathog 2017. [PMID: 28628650 PMCID: PMC5491327 DOI: 10.1371/journal.ppat.1006454] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human Cytomegalovirus (HCMV) is a widespread pathogen, infection with which can cause severe disease for immunocompromised individuals. The complex changes wrought on the host's immune system during both productive and latent HCMV infection are well known. Infected cells are masked and manipulated and uninfected immune cells are also affected; peripheral blood mononuclear cell (PBMC) proliferation is reduced and cytokine profiles altered. Levels increase of the anti-inflammatory cytokine IL-10, which may be important for the establishment of HCMV infections and is required for the development of high viral titres by murine cytomegalovirus. The mechanisms by which HCMV affects T cell IL-10 secretion are not understood. We show here that treatment of PBMC with purified pUL11 induces IL-10 producing T cells as a result of pUL11 binding to the CD45 phosphatase on T cells. IL-10 production induced by HCMV infection is also in part mediated by pUL11. Supernatants from pUL11 treated cells have anti-inflammatory effects on untreated PBMC. Considering the mechanism, CD45 can be a positive or negative regulator of TCR signalling, depending on its expression level, and we show that pUL11 also has concentration dependent activating or inhibitory effects on T cell proliferation and on the kinase function of the CD45 substrate Lck. pUL11 is therefore the first example of a viral protein that can target CD45 to induce T cells with anti-inflammatory properties. It is also the first HCMV protein shown to induce T cell IL-10 secretion. Understanding the mechanisms by which pUL11-induced changes in signal strength influence T cell development and function may provide the basis for the development of novel antiviral treatments and therapies against immune pathologies.
Collapse
Affiliation(s)
- Jasmin Zischke
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
| | - Panagiota Mamareli
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Claudia Pokoyski
- Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Ildar Gabaev
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Sabine Buyny
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Roland Jacobs
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Christine S. Falk
- German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - Matthias Lochner
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Thomas F. Schulz
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
| | - Penelope C. Kay-Fedorov
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
- * E-mail:
| |
Collapse
|
26
|
Thiel N, Keyser KA, Lemmermann NAW, Oduro JD, Wagner K, Elsner C, Halenius A, Lenac Roviš T, Brinkmann MM, Jonjić S, Cicin-Sain L, Messerle M. The Mouse Cytomegalovirus Gene m42 Targets Surface Expression of the Protein Tyrosine Phosphatase CD45 in Infected Macrophages. PLoS Pathog 2016; 12:e1006057. [PMID: 27926943 PMCID: PMC5142792 DOI: 10.1371/journal.ppat.1006057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 11/11/2016] [Indexed: 01/28/2023] Open
Abstract
The receptor-like protein tyrosine phosphatase CD45 is expressed on the surface of cells of hematopoietic origin and has a pivotal role for the function of these cells in the immune response. Here we report that following infection of macrophages with mouse cytomegalovirus (MCMV) the cell surface expression of CD45 is drastically diminished. Screening of a set of MCMV deletion mutants allowed us to identify the viral gene m42 of being responsible for CD45 down-modulation. Moreover, expression of m42 independent of viral infection upon retroviral transduction of the RAW264.7 macrophage cell line led to comparable regulation of CD45 expression. In immunocompetent mice infected with an m42 deletion mutant lower viral titers were observed in all tissues examined when compared to wildtype MCMV, indicating an important role of m42 for viral replication in vivo. The m42 gene product was identified as an 18 kDa protein expressed with early kinetics and is predicted to be a tail-anchored membrane protein. Tracking of surface-resident CD45 molecules revealed that m42 induces internalization and degradation of CD45. The observation that the amounts of the E3 ubiquitin ligases Itch and Nedd4 were diminished in cells expressing m42 and that disruption of a PY motif in the N-terminal part of m42 resulted in loss of function, suggest that m42 acts as an activator or adaptor for these Nedd4-like ubiquitin ligases, which mark CD45 for lysosomal degradation. In conclusion, the down-modulation of CD45 expression in MCMV-infected myeloid cells represents a novel pathway of virus-host interaction. Human cytomegalovirus (HCMV) is a tenacious pathogen, which can be life-threatening for immunocompromised patients and immunologically immature newborns. The pathogenicity of HCMV is owed to a plethora of immunomodulatory functions that interfere with host defense mechanisms. Such viral functions can teach us about viral pathogenesis mechanisms, and also about the functioning of immune cells. In this study we report that the mouse cytomegalovirus (MCMV)–a close relative of HCMV–influences surface expression of the cellular protein CD45 on macrophages and we identified the viral gene m42 mediating this effect. CD45 has long been known to be essential for the functioning of lymphocytes, however, its role in macrophages is less well understood. Growth analysis of a viral mutant indicated that the m42 gene confers a replication advantage to MCMV in vivo. We found that the m42 protein induces internalization of CD45 from the plasma membrane and degradation in lysosomes—most likely triggered by interaction of m42 with a ubiquitin ligase. In our study we detected a new element in the complex interaction of cytomegaloviruses with host cells, and further investigation into this mechanism may provide us with new insights into the functions of CD45 in myeloid cells.
Collapse
Affiliation(s)
- Nadine Thiel
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Kirsten A Keyser
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Niels A W Lemmermann
- Institute of Virology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | - Karen Wagner
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Carina Elsner
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Anne Halenius
- Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Tihana Lenac Roviš
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Melanie M Brinkmann
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stipan Jonjić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Luka Cicin-Sain
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Martin Messerle
- Institute of Virology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
27
|
Bruno L, Cortese M, Monda G, Gentile M, Calò S, Schiavetti F, Zedda L, Cattaneo E, Piccioli D, Schaefer M, Notomista E, Maione D, Carfì A, Merola M, Uematsu Y. Human cytomegalovirus pUL10 interacts with leukocytes and impairs TCR-mediated T-cell activation. Immunol Cell Biol 2016; 94:849-860. [PMID: 27192938 DOI: 10.1038/icb.2016.49] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 05/11/2016] [Accepted: 05/11/2016] [Indexed: 01/27/2023]
Abstract
Human cytomegalovirus (HCMV) is known to exert suppressive effects on the host immune system through expression of various viral genes, thus directly and indirectly affecting antiviral immunity of the infected individuals. We report here that HCMV UL10 encodes a protein (pUL10) with immunosuppressive properties. UL10 has been classified as a member of the HCMV RL11 gene family. Although pUL10 is known to be dispensable for viral replication in cultured cells, its amino-acid sequence is well conserved among different HCMV isolates, suggesting that the protein has a crucial role in viral survival in the host environment. We show that pUL10 is cleaved from the cell surface of fibroblasts as well as epithelial cells and interacts with a cellular receptor ubiquitously expressed on the surface of human leukocytes, demonstrated by ex vivo cell-based assays and flow cytometric analyses on both lymphoid cell lines and primary blood cells. Furthermore, preincubation of peripheral blood mononuclear cells with purified pUL10 ectodomain results in significantly impaired proliferation and substantially reduced pro-inflammatory cytokine production, in particular in CD4+ T cells upon in vitro T-cell stimulation. The inhibitory effect of pUL10 is also observed on antigen receptor-mediated intracellular tyrosine phosphorylation in a T-cell line. Based on these observations, we suggest that pUL10 is a newly identified immunomodulatory protein encoded by HCMV. Further elucidation of interactions between pUL10 and the host immune system during HCMV may contribute to finding ways towards new therapies for HCMV infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Eugenio Notomista
- Department of Biology, University of Naples 'Federico II', Naples, Italy
| | | | | | - Marcello Merola
- Research, GSK Vaccines, Siena, Italy.,Department of Biology, University of Naples 'Federico II', Naples, Italy
| | | |
Collapse
|
28
|
Martinez-Martin N, Ramani SR, Hackney JA, Tom I, Wranik BJ, Chan M, Wu J, Paluch MT, Takeda K, Hass PE, Clark H, Gonzalez LC. The extracellular interactome of the human adenovirus family reveals diverse strategies for immunomodulation. Nat Commun 2016; 7:11473. [PMID: 27145901 PMCID: PMC4858740 DOI: 10.1038/ncomms11473] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 03/30/2016] [Indexed: 01/06/2023] Open
Abstract
Viruses encode secreted and cell-surface expressed proteins essential to modulate host immune defenses and establish productive infections. However, to date there has been no systematic study of the extracellular interactome of any human virus. Here we utilize the E3 proteins, diverse and rapidly evolving transmembrane-containing proteins encoded by human adenoviruses, as a model system to survey the extracellular immunomodulatory landscape. From a large-scale protein interaction screen against a microarray of more than 1,500 human proteins, we find and validate 51 previously unidentified virus–host interactions. Our results uncover conserved strategies as well as substantial diversity and multifunctionality in host targeting within and between viral species. Prominent modulation of the leukocyte immunoglobulin-like and signalling lymphocyte activation molecule families and a number of inhibitory receptors were identified as hubs for viral perturbation, suggesting unrecognized immunoregulatory strategies. We describe a virus–host extracellular interaction map of unprecedented scale that provides new insights into viral immunomodulation. Viruses interact with their hosts via secreted and membrane-bound proteins to affect host immune responses and virulence. Here the authors contribute to our understanding of this relationship with an extracellular interaction map of human and adenoviral E3 immunomodulatory proteins.
Collapse
Affiliation(s)
- Nadia Martinez-Martin
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Sree R Ramani
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Jason A Hackney
- Department of Bioinformatics and Computational Biology, Genentech, 455 East Grand Avenue, South San Francisco, California 94080, USA
| | - Irene Tom
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Bernd J Wranik
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Michelle Chan
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Johnny Wu
- Department of Bioinformatics and Computational Biology, Genentech, 455 East Grand Avenue, South San Francisco, California 94080, USA
| | - Maciej T Paluch
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Kentaro Takeda
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Philip E Hass
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Hilary Clark
- Department of Bioinformatics and Computational Biology, Genentech, 455 East Grand Avenue, South San Francisco, California 94080, USA
| | - Lino C Gonzalez
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| |
Collapse
|
29
|
Windheim M, Höning S, Leppard KN, Butler L, Seed C, Ponnambalam S, Burgert HG. Sorting Motifs in the Cytoplasmic Tail of the Immunomodulatory E3/49K Protein of Species D Adenoviruses Modulate Cell Surface Expression and Ectodomain Shedding. J Biol Chem 2016; 291:6796-812. [PMID: 26841862 DOI: 10.1074/jbc.m115.684787] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Indexed: 11/06/2022] Open
Abstract
The E3 transcription unit of human species C adenoviruses (Ads) encodes immunomodulatory proteins that mediate direct protection of infected cells. Recently, we described a novel immunomodulatory function for E3/49K, an E3 protein uniquely expressed by species D Ads. E3/49K of Ad19a/Ad64, a serotype that causes epidemic keratokonjunctivitis, is synthesized as a highly glycosylated type I transmembrane protein that is subsequently cleaved, resulting in secretion of its large ectodomain (sec49K). sec49K binds to CD45 on leukocytes, impairing activation and functions of natural killer cells and T cells. E3/49K is localized in the Golgi/trans-Golgi network (TGN), in the early endosomes, and on the plasma membrane, yet the cellular compartment where E3/49K is cleaved and the protease involved remained elusive. Here we show that TGN-localized E3/49K comprises both newly synthesized and recycled molecules. Full-length E3/49K was not detected in late endosomes/lysosomes, but the C-terminal fragment accumulated in this compartment at late times of infection. Inhibitor studies showed that cleavage occurs in a post-TGN compartment and that lysosomotropic agents enhance secretion. Interestingly, the cytoplasmic tail of E3/49K contains two potential sorting motifs, YXXΦ (where Φ represents a bulky hydrophobic amino acid) and LL, that are important for binding the clathrin adaptor proteins AP-1 and AP-2in vitro Surprisingly, mutating the LL motif, either alone or together with YXXΦ, did not prevent proteolytic processing but increased cell surface expression and secretion. Upon brefeldin A treatment, cell surface expression was rapidly lost, even for mutants lacking all known endocytosis motifs. Together with immunofluorescence data, we propose a model for intracellular E3/49K transport whereby cleavage takes place on the cell surface by matrix metalloproteases.
Collapse
Affiliation(s)
- Mark Windheim
- From the School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom, the Institute of Biochemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Stefan Höning
- the Institute for Biochemistry I and Center for Molecular Medicine Cologne, 50931 Cologne, Germany, and
| | - Keith N Leppard
- From the School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Larissa Butler
- From the School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Christina Seed
- From the School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Sreenivasan Ponnambalam
- the School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Hans-Gerhard Burgert
- From the School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom,
| |
Collapse
|
30
|
Lassalle F, Depledge DP, Reeves MB, Brown AC, Christiansen MT, Tutill HJ, Williams RJ, Einer-Jensen K, Holdstock J, Atkinson C, Brown JR, van Loenen FB, Clark DA, Griffiths PD, Verjans GM, Schutten M, Milne RS, Balloux F, Breuer J. Islands of linkage in an ocean of pervasive recombination reveals two-speed evolution of human cytomegalovirus genomes. Virus Evol 2016; 2:vew017. [PMID: 30288299 PMCID: PMC6167919 DOI: 10.1093/ve/vew017] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Human cytomegalovirus (HCMV) infects most of the population worldwide, persisting throughout the host's life in a latent state with periodic episodes of reactivation. While typically asymptomatic, HCMV can cause fatal disease among congenitally infected infants and immunocompromised patients. These clinical issues are compounded by the emergence of antiviral resistance and the absence of an effective vaccine, the development of which is likely complicated by the numerous immune evasins encoded by HCMV to counter the host's adaptive immune responses, a feature that facilitates frequent super-infections. Understanding the evolutionary dynamics of HCMV is essential for the development of effective new drugs and vaccines. By comparing viral genomes from uncultivated or low-passaged clinical samples of diverse origins, we observe evidence of frequent homologous recombination events, both recent and ancient, and no structure of HCMV genetic diversity at the whole-genome scale. Analysis of individual gene-scale loci reveals a striking dichotomy: while most of the genome is highly conserved, recombines essentially freely and has evolved under purifying selection, 21 genes display extreme diversity, structured into distinct genotypes that do not recombine with each other. Most of these hyper-variable genes encode glycoproteins involved in cell entry or escape of host immunity. Evidence that half of them have diverged through episodes of intense positive selection suggests that rapid evolution of hyper-variable loci is likely driven by interactions with host immunity. It appears that this process is enabled by recombination unlinking hyper-variable loci from strongly constrained neighboring sites. It is conceivable that viral mechanisms facilitating super-infection have evolved to promote recombination between diverged genotypes, allowing the virus to continuously diversify at key loci to escape immune detection, while maintaining a genome optimally adapted to its asymptomatic infectious lifecycle.
Collapse
Affiliation(s)
- Florent Lassalle
- UCL Genetics Institute, University College London, London, United Kingdom
| | - Daniel P. Depledge
- Division of Infection and Immunity, University College London, London, United
Kingdom
| | - Matthew B. Reeves
- Division of Infection and Immunity, University College London, London, United
Kingdom
| | | | - Mette T. Christiansen
- Division of Infection and Immunity, University College London, London, United
Kingdom
| | - Helena J. Tutill
- Division of Infection and Immunity, University College London, London, United
Kingdom
| | - Rachel J. Williams
- Division of Infection and Immunity, University College London, London, United
Kingdom
| | | | | | - Claire Atkinson
- Department of Virology, Royal Free Hospital, London, United Kingdom
| | - Julianne R. Brown
- Microbiology, Virology and Infection Prevention and Control, Camelia Botnar
Laboratories, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United
Kingdom
| | | | - Duncan A. Clark
- Department of Virology, Barts Health NHS Trust, London, United Kingdom
| | - Paul D. Griffiths
- Division of Infection and Immunity, University College London, London, United
Kingdom
| | | | - Martin Schutten
- Department of Viroscience, Erasmus, MC Rotterdam, the Netherlands
| | - Richard S.B. Milne
- Division of Infection and Immunity, University College London, London, United
Kingdom
| | - Francois Balloux
- UCL Genetics Institute, University College London, London, United Kingdom
| | - Judith Breuer
- Division of Infection and Immunity, University College London, London, United
Kingdom
| |
Collapse
|
31
|
Decoding protein networks during virus entry by quantitative proteomics. Virus Res 2015; 218:25-39. [PMID: 26365680 PMCID: PMC4914609 DOI: 10.1016/j.virusres.2015.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/31/2015] [Accepted: 09/08/2015] [Indexed: 01/05/2023]
Abstract
Virus entry into host cells relies on interactions between viral and host structures including lipids, carbohydrates and proteins. Particularly, protein–protein interactions between viral surface proteins and host proteins as well as secondary host protein–protein interactions play a pivotal role in coordinating virus binding and uptake. These interactions are dynamic and frequently involve multiprotein complexes. In the past decade mass spectrometry based proteomics methods have reached sensitivities and high throughput compatibilities of genomics methods and now allow the reliable quantitation of proteins in complex samples from limited material. As proteomics provides essential information on the biologically active entity namely the protein, including its posttranslational modifications and its interactions with other proteins, it is an indispensable method in the virologist's toolbox. Here we review protein interactions during virus entry and compare classical biochemical methods to study entry with novel technically advanced quantitative proteomics techniques. We highlight the value of quantitative proteomics in mapping functional virus entry networks, discuss the benefits and limitations and illustrate how the methodology will help resolve unsettled questions in virus entry research in the future.
Collapse
|
32
|
High-throughput analysis of human cytomegalovirus genome diversity highlights the widespread occurrence of gene-disrupting mutations and pervasive recombination. J Virol 2015; 89:7673-7695. [PMID: 25972543 DOI: 10.1128/jvi.00578-15] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Human cytomegalovirus is a widespread pathogen of major medical importance. It causes significant morbidity and mortality in the immunocompromised and congenital infections can result in severe disabilities or stillbirth. Development of a vaccine is prioritized, but no candidate is close to release. Although correlations of viral genetic variability with pathogenicity are suspected, knowledge about strain diversity of the 235kb genome is still limited. In this study, 96 full-length human cytomegalovirus genomes from clinical isolates were characterized, quadrupling the available information for full-genome analysis. These data provide the first high-resolution map of human cytomegalovirus interhost diversity and evolution. We show that cytomegalovirus is significantly more divergent than all other human herpesviruses and highlight hotspots of diversity in the genome. Importantly, 75% of strains are not genetically intact, but contain disruptive mutations in a diverse set of 26 genes, including immunomodulative genes UL40 and UL111A. These mutants are independent from culture passaging artifacts and circulate in natural populations. Pervasive recombination, which is linked to the widespread occurrence of multiple infections, was found throughout the genome. Recombination density was significantly higher than in other human herpesviruses and correlated with strain diversity. While the overall effects of strong purifying selection on virus evolution are apparent, evidence of diversifying selection was found in several genes encoding proteins that interact with the host immune system, including UL18, UL40, UL142 and UL147. These residues may present phylogenetic signatures of past and ongoing virus-host interactions. IMPORTANCE Human cytomegalovirus has the largest genome of all viruses that infect humans. Currently, there is a great interest in establishing associations between genetic variants and strain pathogenicity of this herpesvirus. Since the number of publicly available full-genome sequences is limited, knowledge about strain diversity is highly fragmented and biased towards a small set of loci. Combined with our previous work, we have now contributed 101 complete genome sequences. We have used these data to conduct the first high-resolution analysis of interhost genome diversity, providing an unbiased and comprehensive overview of cytomegalovirus variability. These data are of major value to the development of novel antivirals and a vaccine and to identify potential targets for genotype-phenotype experiments. Furthermore, they have enabled a thorough study of the evolutionary processes that have shaped cytomegalovirus diversity.
Collapse
|
33
|
Viral interference with functions of the cellular receptor tyrosine phosphatase CD45. Viruses 2015; 7:1540-57. [PMID: 25807057 PMCID: PMC4379584 DOI: 10.3390/v7031540] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 12/24/2022] Open
Abstract
The receptor tyrosine phosphatase CD45 is expressed on the surface of almost all cells of hematopoietic origin. CD45 functions are central to the development of T cells and determine the threshold at which T and B lymphocytes can become activated. Given this pivotal role of CD45 in the immune system, it is probably not surprising that viruses interfere with the activity of CD45 in lymphocytes to dampen the immune response and that they also utilize this molecule to accomplish their replication cycle. Here we report what is known about the interaction of viral proteins with CD45. Moreover, we debate putative interactions of viruses with CD45 in myeloid cells and the resulting consequences-subjects that remain to be investigated. Finally, we summarize the evidence that pathogens were the driving force for the evolution of CD45.
Collapse
|
34
|
Brinkmann MM, Dağ F, Hengel H, Messerle M, Kalinke U, Čičin-Šain L. Cytomegalovirus immune evasion of myeloid lineage cells. Med Microbiol Immunol 2015; 204:367-82. [PMID: 25776081 DOI: 10.1007/s00430-015-0403-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 02/28/2015] [Indexed: 12/23/2022]
Abstract
Cytomegalovirus (CMV) evades the immune system in many different ways, allowing the virus to grow and its progeny to spread in the face of an adverse environment. Mounting evidence about the antiviral role of myeloid immune cells has prompted the research of CMV immune evasion mechanisms targeting these cells. Several cells of the myeloid lineage, such as monocytes, dendritic cells and macrophages, play a role in viral control, but are also permissive for CMV and are naturally infected by it. Therefore, CMV evasion of myeloid cells involves mechanisms that qualitatively differ from the evasion of non-CMV-permissive immune cells of the lymphoid lineage. The evasion of myeloid cells includes effects in cis, where the virus modulates the immune signaling pathways within the infected myeloid cell, and those in trans, where the virus affects somatic cells targeted by cytokines released from myeloid cells. This review presents an overview of CMV strategies to modulate and evade the antiviral activity of myeloid cells in cis and in trans.
Collapse
Affiliation(s)
- Melanie M Brinkmann
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Brunswick, Germany
| | | | | | | | | | | |
Collapse
|
35
|
ORF57 overcomes the detrimental sequence bias of Kaposi's sarcoma-associated herpesvirus lytic genes. J Virol 2015; 89:5097-109. [PMID: 25694606 DOI: 10.1128/jvi.03264-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/17/2015] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Kaposi's sarcoma-associated herpesvirus (KSHV) encodes ORF57, which enhances the expression of intronless KSHV genes on multiple posttranscriptional levels. However, it remains elusive how ORF57 recognizes viral RNAs. Here, we demonstrate that ORF57 also increases the expression of the multiple intron-containing K15 gene. The nucleotide bias of the K15 cDNA revealed an unusual high AT content. Thus, we optimized the K15 cDNA by raising the frequency of GC nucleotides, yielding an ORF57-independent version. To further prove the importance of the sequence bias of ORF57-dependent RNAs, we grouped KSHV mRNAs according to their AT content and found a correlation between AT-richness and ORF57 dependency. More importantly, latent genes, which have to be expressed in the absence of ORF57, have a low AT content and are indeed ORF57 independent. The nucleotide composition of K15 resembles that of HIV gag, which cannot be expressed unless RNA export is facilitated by the HIV Rev protein. Interestingly, ORF57 can partially rescue HIV Gag expression. Thus, the KSHV target RNAs of ORF57 and HIV gag RNA may share certain motifs based on the nucleotide bias. A bioinformatic comparison between wild-type and sequence-optimized K15 revealed a higher density for hnRNP-binding motifs in the former. We speculate that binding of particular hnRNPs to KSHV lytic transcripts is the prerequisite for ORF57 to enhance their expression. IMPORTANCE The mostly intronless genes of KSHV are only expressed in the presence of the viral regulator protein ORF57, but how ORF57 recognizes viral RNAs remains elusive. We focused on the multiple intron-containing KSHV gene K15 and revealed that its expression is also increased by ORF57. Moreover, sequences in the K15 cDNA mediate this enhancement. The quest for a target sequence or a response element for ORF57 in the lytic genes was not successful. Instead, we found the nucleotide bias to be the critical determinant of ORF57 dependency. Based on the fact that ORF57 has only a weak affinity for nucleic acids, we speculate that a cellular RNA-binding protein provides the sequence preference for ORF57. This study provides evidence that herpesviral RNA regulator proteins use the sequence bias of lytic genes and the resulting composition of the viral mRNP to distinguish between viral and cellular mRNAs.
Collapse
|
36
|
Gabaev I, Elbasani E, Ameres S, Steinbrück L, Stanton R, Döring M, Lenac Rovis T, Kalinke U, Jonjic S, Moosmann A, Messerle M. Expression of the human cytomegalovirus UL11 glycoprotein in viral infection and evaluation of its effect on virus-specific CD8 T cells. J Virol 2014; 88:14326-39. [PMID: 25275132 PMCID: PMC4249143 DOI: 10.1128/jvi.01691-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 09/25/2014] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The human cytomegalovirus (CMV) UL11 open reading frame (ORF) encodes a putative type I transmembrane glycoprotein which displays remarkable amino acid sequence variability among different CMV isolates, suggesting that it represents an important virulence factor. In a previous study, we have shown that UL11 can interact with the cellular receptor tyrosine phosphatase CD45, which has a central role for signal transduction in T cells, and treatment of T cells with large amounts of a soluble UL11 protein inhibited their proliferation. In order to analyze UL11 expression in CMV-infected cells, we constructed CMV recombinants whose genomes either encode tagged UL11 versions or carry a stop mutation in the UL11 ORF. Moreover, we examined whether UL11 affects the function of virus-specific cytotoxic T lymphocytes (CTLs). We found that the UL11 ORF gives rise to several proteins due to both posttranslational modification and alternative translation initiation sites. Biotin labeling of surface proteins on infected cells indicated that only highly glycosylated UL11 forms are present at the plasma membrane, whereas less glycosylated UL11 forms were found in the endoplasmic reticulum. We did not find evidence of UL11 cleavage or secretion of a soluble UL11 version. Cocultivation of CTLs recognizing different CMV epitopes with fibroblasts infected with a UL11 deletion mutant or the parental strain revealed that under the conditions applied UL11 did not influence the activation of CMV-specific CD8 T cells. For further studies, we propose to investigate the interaction of UL11 with CD45 and the functional consequences in other immune cells expressing CD45. IMPORTANCE Human cytomegalovirus (CMV) belongs to those viruses that extensively interfere with the host immune response, yet the precise function of many putative immunomodulatory CMV proteins remains elusive. Previously, we have shown that the CMV UL11 protein interacts with the leukocyte common antigen CD45, a cellular receptor tyrosine phosphatase with a central role for signal transduction in T cells. Here, we examined the proteins expressed by the UL11 gene in CMV-infected cells and found that at least one form of UL11 is present at the cell surface, enabling it to interact with CD45 on immune cells. Surprisingly, CMV-expressed UL11 did not affect the activity of virus-specific CD8 T cells. This finding warrants investigation of the impact of UL11 on CD45 functions in other leukocyte subpopulations.
Collapse
Affiliation(s)
- Ildar Gabaev
- Department of Virology, Hannover Medical School, Hannover, Germany
| | - Endrit Elbasani
- Department of Virology, Hannover Medical School, Hannover, Germany
| | - Stefanie Ameres
- Clinical Cooperation Group Immunooncology, Helmholtz Centre Munich, Munich, Germany
| | - Lars Steinbrück
- Department of Virology, Hannover Medical School, Hannover, Germany
| | - Richard Stanton
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Marius Döring
- Centre for Experimental and Clinical Infection Research, Twincore, Hannover, Germany
| | - Tihana Lenac Rovis
- Department of Histology and Embryology and Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Ulrich Kalinke
- Centre for Experimental and Clinical Infection Research, Twincore, Hannover, Germany
| | - Stipan Jonjic
- Department of Histology and Embryology and Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Andreas Moosmann
- Clinical Cooperation Group Immunooncology, Helmholtz Centre Munich, Munich, Germany German Center for Infection Research (DZIF), partner sites, Hannover and Munich, Germany
| | - Martin Messerle
- Department of Virology, Hannover Medical School, Hannover, Germany German Center for Infection Research (DZIF), partner sites, Hannover and Munich, Germany
| |
Collapse
|
37
|
Jean Beltran PM, Cristea IM. The life cycle and pathogenesis of human cytomegalovirus infection: lessons from proteomics. Expert Rev Proteomics 2014; 11:697-711. [PMID: 25327590 DOI: 10.1586/14789450.2014.971116] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Viruses have coevolved with their hosts, acquiring strategies to subvert host cellular pathways for effective viral replication and spread. Human cytomegalovirus (HCMV), a widely-spread β-herpesvirus, is a major cause of birth defects and opportunistic infections in HIV-1/AIDS patients. HCMV displays an intricate system-wide modulation of the human cell proteome. An impressive array of virus-host protein interactions occurs throughout the infection. To investigate the virus life cycle, proteomics has recently become a significant component of virology studies. Here, we review the mass spectrometry-based proteomics approaches used in HCMV studies, as well as their contribution to understanding the HCMV life cycle and the virus-induced changes to host cells. The importance of the biological insights gained from these studies clearly demonstrate the impact that proteomics has had and can continue to have on understanding HCMV biology and identifying new therapeutic targets.
Collapse
Affiliation(s)
- Pierre M Jean Beltran
- Department of Molecular Biology, 210 Lewis Thomas Laboratory, Princeton University, Princeton, New Jersey, NJ 08544, USA
| | | |
Collapse
|
38
|
Weekes MP, Tomasec P, Huttlin EL, Fielding CA, Nusinow D, Stanton RJ, Wang ECY, Aicheler R, Murrell I, Wilkinson GWG, Lehner PJ, Gygi SP. Quantitative temporal viromics: an approach to investigate host-pathogen interaction. Cell 2014; 157:1460-1472. [PMID: 24906157 PMCID: PMC4048463 DOI: 10.1016/j.cell.2014.04.028] [Citation(s) in RCA: 345] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 02/18/2014] [Accepted: 04/03/2014] [Indexed: 12/11/2022]
Abstract
A systematic quantitative analysis of temporal changes in host and viral proteins throughout the course of a productive infection could provide dynamic insights into virus-host interaction. We developed a proteomic technique called “quantitative temporal viromics” (QTV), which employs multiplexed tandem-mass-tag-based mass spectrometry. Human cytomegalovirus (HCMV) is not only an important pathogen but a paradigm of viral immune evasion. QTV detailed how HCMV orchestrates the expression of >8,000 cellular proteins, including 1,200 cell-surface proteins to manipulate signaling pathways and counterintrinsic, innate, and adaptive immune defenses. QTV predicted natural killer and T cell ligands, as well as 29 viral proteins present at the cell surface, potential therapeutic targets. Temporal profiles of >80% of HCMV canonical genes and 14 noncanonical HCMV open reading frames were defined. QTV is a powerful method that can yield important insights into viral infection and is applicable to any virus with a robust in vitro model. PaperClip
>8,000 proteins quantified over eight time points, including 1,200 cell-surface proteins Temporal profiles of 139/171 canonical HCMV proteins and 14 noncanonical HCMV ORFs Multiple families of cell-surface receptors selectively modulated by HCMV Multiple signaling pathways modulated during HCMV infection
Collapse
Affiliation(s)
- Michael P Weekes
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA; Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK.
| | - Peter Tomasec
- School of Medicine, Cardiff University, Tenovus Building, Heath Park, Cardiff CF14 4XX, UK
| | - Edward L Huttlin
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Ceri A Fielding
- School of Medicine, Cardiff University, Tenovus Building, Heath Park, Cardiff CF14 4XX, UK
| | - David Nusinow
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Richard J Stanton
- School of Medicine, Cardiff University, Tenovus Building, Heath Park, Cardiff CF14 4XX, UK
| | - Eddie C Y Wang
- School of Medicine, Cardiff University, Tenovus Building, Heath Park, Cardiff CF14 4XX, UK
| | - Rebecca Aicheler
- School of Medicine, Cardiff University, Tenovus Building, Heath Park, Cardiff CF14 4XX, UK
| | - Isa Murrell
- School of Medicine, Cardiff University, Tenovus Building, Heath Park, Cardiff CF14 4XX, UK
| | - Gavin W G Wilkinson
- School of Medicine, Cardiff University, Tenovus Building, Heath Park, Cardiff CF14 4XX, UK
| | - Paul J Lehner
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
39
|
Greco TM, Diner BA, Cristea IM. The Impact of Mass Spectrometry-Based Proteomics on Fundamental Discoveries in Virology. Annu Rev Virol 2014; 1:581-604. [PMID: 26958735 DOI: 10.1146/annurev-virology-031413-085527] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In recent years, mass spectrometry has emerged as a core component of fundamental discoveries in virology. As a consequence of their coevolution, viruses and host cells have established complex, dynamic interactions that function either in promoting virus replication and dissemination or in host defense against invading pathogens. Thus, viral infection triggers an impressive range of proteome changes. Alterations in protein abundances, interactions, posttranslational modifications, subcellular localizations, and secretion are temporally regulated during the progression of an infection. Consequently, understanding viral infection at the molecular level requires versatile approaches that afford both breadth and depth of analysis. Mass spectrometry is uniquely positioned to bridge this experimental dichotomy. Its application to both unbiased systems analyses and targeted, hypothesis-driven studies has accelerated discoveries in viral pathogenesis and host defense. Here, we review the contributions of mass spectrometry-based proteomic approaches to understanding viral morphogenesis, replication, and assembly and to characterizing host responses to infection.
Collapse
Affiliation(s)
- Todd M Greco
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544;
| | - Benjamin A Diner
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544;
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544;
| |
Collapse
|
40
|
Van Damme E, Van Loock M. Functional annotation of human cytomegalovirus gene products: an update. Front Microbiol 2014; 5:218. [PMID: 24904534 PMCID: PMC4032930 DOI: 10.3389/fmicb.2014.00218] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/25/2014] [Indexed: 01/31/2023] Open
Abstract
Human cytomegalovirus is an opportunistic double-stranded DNA virus with one of the largest viral genomes known. The 235 kB genome is divided in a unique long (UL) and a unique short (US) region which are flanked by terminal and internal repeats. The expression of HCMV genes is highly complex and involves the production of protein coding transcripts, polyadenylated long non-coding RNAs, polyadenylated anti-sense transcripts and a variety of non-polyadenylated RNAs such as microRNAs. Although the function of many of these transcripts is unknown, they are suggested to play a direct or regulatory role in the delicately orchestrated processes that ensure HCMV replication and life-long persistence. This review focuses on annotating the complete viral genome based on three sources of information. First, previous reviews were used as a template for the functional keywords to ensure continuity; second, the Uniprot database was used to further enrich the functional database; and finally, the literature was manually curated for novel functions of HCMV gene products. Novel discoveries were discussed in light of the viral life cycle. This functional annotation highlights still poorly understood regions of the genome but more importantly it can give insight in functional clusters and/or may be helpful in the analysis of future transcriptomics and proteomics studies.
Collapse
Affiliation(s)
- Ellen Van Damme
- Janssen Infectious Diseases BVBA, Therapeutic Area of Infectious Diseases Beerse, Belgium
| | - Marnix Van Loock
- Janssen Infectious Diseases BVBA, Therapeutic Area of Infectious Diseases Beerse, Belgium
| |
Collapse
|
41
|
Hu C, Chen J, Ye L, Chen R, Zhang L, Xue X. Codon usage bias in human cytomegalovirus and its biological implication. Gene 2014; 545:5-14. [PMID: 24814188 DOI: 10.1016/j.gene.2014.05.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 10/25/2022]
Abstract
Human cytomegalovirus (HCMV) infection, a worldwide contagion, causes a serious disorder in infected individuals. Analysis of codon usage can reveal much molecular information about this virus. The effective number of codon (ENC) values, relative synonymous codon usage (RSCU) values, codon adaptation index (CAI), and nucleotide contents was investigated in approximately 160 coding sequences (CDS) among 17 human cytomegalovirus genomes using the software CodonW. Linear regression analysis and logistic regression were performed to explore the preliminary data. The results showed that, overall, HCMV genomes had low codon usage bias (mean ENC=47.619). However, the ENC of individual CDS varied widely and was distributed unevenly between host-related genes and viral-self-function genes (P=0.002, odds ratio (OR)=3.194), as did the GC content (P=0.016, OR=2.178). The ENC values correlated with CAI, GC content, and the nucleotide composing at the 3rd codon position (GC3s) (P<0.001). There was a significant variation in the codon preference that depended on the RSCU data. The predicted ENC curve suggested that mutational pressure, rather than natural selection, was one of the main factors that determined the codon usage bias in HCMV. Among 123 genes with known function, the genes related to viral self-replication and viral-host interaction showed different ENC and CAI values, and GC and GC3s contents. In conclusion, the detailed codon usage bias theoretically revealed information concerning HCMV evolution and could be a valuable additional parameter for HCMV gene function research.
Collapse
Affiliation(s)
- Changyuan Hu
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Ouhai District 325035, Wenzhou City, Zhejiang Province, China
| | - Jing Chen
- Department of Rheumatism and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Ouhai District 325035, Wenzhou City, Zhejiang Province, China
| | - Lulu Ye
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Ouhai District 325035, Wenzhou City, Zhejiang Province, China
| | - Renpin Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Ouhai District 325035, Wenzhou City, Zhejiang Province, China
| | - Lifang Zhang
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Ouhai District 325035, Wenzhou City, Zhejiang Province, China
| | - Xiangyang Xue
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Ouhai District 325035, Wenzhou City, Zhejiang Province, China.
| |
Collapse
|
42
|
Fielding CA, Aicheler R, Stanton RJ, Wang ECY, Han S, Seirafian S, Davies J, McSharry BP, Weekes MP, Antrobus PR, Prod'homme V, Blanchet FP, Sugrue D, Cuff S, Roberts D, Davison AJ, Lehner PJ, Wilkinson GWG, Tomasec P. Two novel human cytomegalovirus NK cell evasion functions target MICA for lysosomal degradation. PLoS Pathog 2014; 10:e1004058. [PMID: 24787765 PMCID: PMC4006889 DOI: 10.1371/journal.ppat.1004058] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 02/18/2014] [Indexed: 02/07/2023] Open
Abstract
NKG2D plays a major role in controlling immune responses through the regulation of natural killer (NK) cells, αβ and γδ T-cell function. This activating receptor recognizes eight distinct ligands (the MHC Class I polypeptide-related sequences (MIC) A andB, and UL16-binding proteins (ULBP)1-6) induced by cellular stress to promote recognition cells perturbed by malignant transformation or microbial infection. Studies into human cytomegalovirus (HCMV) have aided both the identification and characterization of NKG2D ligands (NKG2DLs). HCMV immediate early (IE) gene up regulates NKGDLs, and we now describe the differential activation of ULBP2 and MICA/B by IE1 and IE2 respectively. Despite activation by IE functions, HCMV effectively suppressed cell surface expression of NKGDLs through both the early and late phases of infection. The immune evasion functions UL16, UL142, and microRNA(miR)-UL112 are known to target NKG2DLs. While infection with a UL16 deletion mutant caused the expected increase in MICB and ULBP2 cell surface expression, deletion of UL142 did not have a similar impact on its target, MICA. We therefore performed a systematic screen of the viral genome to search of addition functions that targeted MICA. US18 and US20 were identified as novel NK cell evasion functions capable of acting independently to promote MICA degradation by lysosomal degradation. The most dramatic effect on MICA expression was achieved when US18 and US20 acted in concert. US18 and US20 are the first members of the US12 gene family to have been assigned a function. The US12 family has 10 members encoded sequentially through US12-US21; a genetic arrangement, which is suggestive of an 'accordion' expansion of an ancestral gene in response to a selective pressure. This expansion must have be an ancient event as the whole family is conserved across simian cytomegaloviruses from old world monkeys. The evolutionary benefit bestowed by the combinatorial effect of US18 and US20 on MICA may have contributed to sustaining the US12 gene family.
Collapse
Affiliation(s)
- Ceri A. Fielding
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Rebecca Aicheler
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Richard J. Stanton
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Eddie C. Y. Wang
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Song Han
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sepehr Seirafian
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - James Davies
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Brian P. McSharry
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael P. Weekes
- Cambridge Institute for Medical Research (CIMR), Wellcome Trust/MRC Building, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - P. Robin Antrobus
- Cambridge Institute for Medical Research (CIMR), Wellcome Trust/MRC Building, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Virginie Prod'homme
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Fabien P. Blanchet
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Daniel Sugrue
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Simone Cuff
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Dawn Roberts
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Andrew J. Davison
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Paul J. Lehner
- Cambridge Institute for Medical Research (CIMR), Wellcome Trust/MRC Building, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Gavin W. G. Wilkinson
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
- * E-mail:
| | - Peter Tomasec
- Section of Medical Microbiology, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
43
|
Alzhanova D, Hammarlund E, Reed J, Meermeier E, Rawlings S, Ray CA, Edwards DM, Bimber B, Legasse A, Planer S, Sprague J, Axthelm MK, Pickup DJ, Lewinsohn DM, Gold MC, Wong SW, Sacha JB, Slifka MK, Früh K. T cell inactivation by poxviral B22 family proteins increases viral virulence. PLoS Pathog 2014; 10:e1004123. [PMID: 24832205 PMCID: PMC4022744 DOI: 10.1371/journal.ppat.1004123] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 04/02/2014] [Indexed: 11/19/2022] Open
Abstract
Infections with monkeypox, cowpox and weaponized variola virus remain a threat to the increasingly unvaccinated human population, but little is known about their mechanisms of virulence and immune evasion. We now demonstrate that B22 proteins, encoded by the largest genes of these viruses, render human T cells unresponsive to stimulation of the T cell receptor by MHC-dependent antigen presentation or by MHC-independent stimulation. In contrast, stimuli that bypass TCR-signaling are not inhibited. In a non-human primate model of monkeypox, virus lacking the B22R homologue (MPXVΔ197) caused only mild disease with lower viremia and cutaneous pox lesions compared to wild type MPXV which caused high viremia, morbidity and mortality. Since MPXVΔ197-infected animals displayed accelerated T cell responses and less T cell dysregulation than MPXV US2003, we conclude that B22 family proteins cause viral virulence by suppressing T cell control of viral dissemination.
Collapse
Affiliation(s)
- Dina Alzhanova
- Vaccine and Gene Therapy Institute, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Erika Hammarlund
- Division of Neuroscience, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Jason Reed
- Vaccine and Gene Therapy Institute, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Erin Meermeier
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
- Portland Veterans Administration Medical Center, Portland, Oregon, United States of America
| | - Stephanie Rawlings
- Vaccine and Gene Therapy Institute, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Caroline A. Ray
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David M. Edwards
- Vaccine and Gene Therapy Institute, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Ben Bimber
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Alfred Legasse
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Shannon Planer
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Jerald Sprague
- Vaccine and Gene Therapy Institute, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Michael K. Axthelm
- Vaccine and Gene Therapy Institute, Oregon National Primate Research Center, Portland, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - David J. Pickup
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David M. Lewinsohn
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
- Portland Veterans Administration Medical Center, Portland, Oregon, United States of America
| | - Marielle C. Gold
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
- Portland Veterans Administration Medical Center, Portland, Oregon, United States of America
| | - Scott W. Wong
- Vaccine and Gene Therapy Institute, Oregon National Primate Research Center, Portland, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Jonah B. Sacha
- Vaccine and Gene Therapy Institute, Oregon National Primate Research Center, Portland, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Mark K. Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Portland, Oregon, United States of America
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon National Primate Research Center, Portland, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Portland, Oregon, United States of America
| |
Collapse
|
44
|
Human cytomegalovirus and autoimmune disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:472978. [PMID: 24967373 PMCID: PMC4022258 DOI: 10.1155/2014/472978] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 03/17/2014] [Indexed: 11/28/2022]
Abstract
Human cytomegalovirus (HCMV) represents a prototypic pathogenic member of the β-subgroup of the herpesvirus family. A range of HCMV features like its lytic replication in multiple tissues, the lifelong persistence through periods of latency and intermitting reactivation, the extraordinary large proteome, and extensive manipulation of adaptive and innate immunity make HCMV a high profile candidate for involvement in autoimmune disorders. We surveyed the available literature for reports on HCMV association with onset or exacerbation of autoimmune disease. A causative linkage between HCMV and systemic lupus erythematosus (SLE), systemic sclerosis (SSc), diabetes mellitus type 1, and rheumatoid arthritis (RA) is suggested by the literature. However, a clear association of HCMV seroprevalence and disease could not be established, leaving the question open whether HCMV could play a coresponsible role for onset of disease. For convincing conclusions population-based prospective studies must be performed in the future. Specific immunopathogenic mechanisms by which HCMV could contribute to the course of autoimmune disease have been suggested, for example, molecular mimicry by UL94 in SSc and UL83/pp65 in SLE patients, as well as aggravation of joint inflammation by induction and expansion of CD4+/CD28− T-cells in RA patients. Further studies are needed to validate these findings and to lay the grounds for targeted therapeutic intervention.
Collapse
|
45
|
Sijmons S, Van Ranst M, Maes P. Genomic and functional characteristics of human cytomegalovirus revealed by next-generation sequencing. Viruses 2014; 6:1049-72. [PMID: 24603756 PMCID: PMC3970138 DOI: 10.3390/v6031049] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 02/11/2014] [Accepted: 02/11/2014] [Indexed: 01/08/2023] Open
Abstract
The complete genome of human cytomegalovirus (HCMV) was elucidated almost 25 years ago using a traditional cloning and Sanger sequencing approach. Analysis of the genetic content of additional laboratory and clinical isolates has lead to a better, albeit still incomplete, definition of the coding potential and diversity of wild-type HCMV strains. The introduction of a new generation of massively parallel sequencing technologies, collectively called next-generation sequencing, has profoundly increased the throughput and resolution of the genomics field. These increased possibilities are already leading to a better understanding of the circulating diversity of HCMV clinical isolates. The higher resolution of next-generation sequencing provides new opportunities in the study of intrahost viral population structures. Furthermore, deep sequencing enables novel diagnostic applications for sensitive drug resistance mutation detection. RNA-seq applications have changed the picture of the HCMV transcriptome, which resulted in proof of a vast amount of splicing events and alternative transcripts. This review discusses the application of next-generation sequencing technologies, which has provided a clearer picture of the intricate nature of the HCMV genome. The continuing development and application of novel sequencing technologies will further augment our understanding of this ubiquitous, but elusive, herpesvirus.
Collapse
Affiliation(s)
- Steven Sijmons
- Laboratory of Clinical Virology, Rega Institute for Medical Research, K.U.Leuven, Minderbroedersstraat 10, Leuven BE-3000, Belgium.
| | - Marc Van Ranst
- Laboratory of Clinical Virology, Rega Institute for Medical Research, K.U.Leuven, Minderbroedersstraat 10, Leuven BE-3000, Belgium.
| | - Piet Maes
- Laboratory of Clinical Virology, Rega Institute for Medical Research, K.U.Leuven, Minderbroedersstraat 10, Leuven BE-3000, Belgium.
| |
Collapse
|
46
|
Qadis AQ, Goya S, Yatsu M, Kimura A, Ichijo T, Sato S. Immune-stimulatory effects of a bacteria-based probiotic on peripheral leukocyte subpopulations and cytokine mRNA expression levels in scouring holstein calves. J Vet Med Sci 2014; 76:189-95. [PMID: 24131856 PMCID: PMC3982805 DOI: 10.1292/jvms.13-0370] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Subpopulations of peripheral leukocytes and cytokine mRNA expression levels were evaluated in scouring and healthy Holstein calves (age 10 ± 5 days; n=42) treated with a probiotic consisting of Lactobacillus plantarum, Enterococcus faecium and Clostridium butyricum. The calves were assigned to the scouring or healthy group and then subdivided into pathogen-positive treated (n=8), pathogen-positive control (n=8), pathogen-negative treated (n=6), pathogen-negative control (n=6), healthy treated (n=6) and healthy control (n=8) groups. A single dose of the probiotic (3.0 g/100 kg body weight) was given to each calf in the treatment groups for 5 days. Blood samples were collected on the first day of scour occurrence (day 0) and on day 7. In the scouring calves, smaller peripheral leukocyte subpopulations and cytokine mRNA expression levels were noted on day 0. The numbers of CD3(+) T cells; CD4(+), CD8(+) and WC1(+) γδ T cell subsets; and CD14(+), CD21(+) and CD282(+) (TLR2) cells were significantly increased in the scouring and healthy treated calves on day 7. Furthermore, interleukin-6, tumor necrosis factor-alpha and interferon-gamma mRNA expression was elevated in the peripheral leukocytes of the scouring and healthy treated calves on day 7. The scouring calves given the probiotic recovered on day 7. A significantly smaller number of peripheral leukocytes and lower cytokine mRNA expression level might be induced by scouring in calves. Repeated probiotic administration might stimulate cellular immunity and encourage recovery from scouring in pre-weaning Holstein calves.
Collapse
Affiliation(s)
- Abdul Qadir Qadis
- United Graduate School of Veterinary Science, Gifu University, Gifu 501-1193, Japan
| | | | | | | | | | | |
Collapse
|
47
|
A unique secreted adenovirus E3 protein binds to the leukocyte common antigen CD45 and modulates leukocyte functions. Proc Natl Acad Sci U S A 2013; 110:E4884-93. [PMID: 24218549 DOI: 10.1073/pnas.1312420110] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The E3 transcription unit of human adenoviruses (Ads) encodes immunomodulatory proteins. Interestingly, the size and composition of the E3 region differs considerably among Ad species, suggesting that distinct sets of immunomodulatory E3 proteins may influence their interaction with the human host and the disease pattern. However, to date, only common immune evasion functions of species C E3 proteins have been described. Here we report on the immunomodulatory activity of a species D-specific E3 protein, E3/49K. Unlike all other E3 proteins that act on infected cells, E3/49K seems to target uninfected cells. Initially synthesized as an 80- to 100-kDa type I transmembrane protein, E3/49K is subsequently cleaved, with the large ectodomain (sec49K) secreted. We found that purified sec49K exhibits specific binding to lymphoid cell lines and all primary leukocytes, but not to fibroblasts or epithelial cells. Consistent with this binding profile and the molecular mass, the sec49K receptor was identified as the cell surface protein tyrosine phosphatase CD45. Antibody-blocking studies suggested that sec49K binds to the membrane proximal domains present in all CD45 isoforms. Functional studies showed that sec49K can suppress the activation and cytotoxicity of natural killer cells as well as the activation, signaling, and cytokine production of T cells. Thus, we have discovered an adenovirus protein that is actively secreted and describe immunomodulatory activities of an E3 protein uniquely expressed by a single Ad species.
Collapse
|
48
|
Prinz I, Thamm K, Port M, Weissinger EM, Stadler M, Gabaev I, Jacobs R, Ganser A, Koenecke C. Donor Vδ1+ γδ T cells expand after allogeneic hematopoietic stem cell transplantation and show reactivity against CMV-infected cells but not against progressing B-CLL. Exp Hematol Oncol 2013; 2:14. [PMID: 23663319 PMCID: PMC3669054 DOI: 10.1186/2162-3619-2-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 05/10/2013] [Indexed: 11/30/2022] Open
Abstract
γδ T lymphocytes play an important role in immune reactions towards infections and malignancies. In particular, Vγ9–Vδ1+ T lymphocytes are thought to play protective antiviral roles in human CMV infection. Recently, Vδ1+ T lymphocytes were proposed to also have anti- B-CLL reactivity. Here we report a case of 48-year-old man who received allogeneic stem cell transplantation for progressive B-CLL. Within one year after transplantation, lymphoma relapsed despite a dramatic increase of Vδ1+ T cells in the patient’s blood. In vitro killing assays revealed activity of patient’s γδ cells against CMV target cells, but not against the relapsing lymphoma-cells. This argues for a contribution of Vδ1+ cells in the immune reaction against CMV reactivation, but does not support a strong correlation of expanded Vδ1+ T cells and favorable disease outcome in B-CLL patients.
Collapse
Affiliation(s)
- Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Fornara O, Odeberg J, Khan Z, Stragliotto G, Peredo I, Butler L, Söderberg-Nauclér C. Human cytomegalovirus particles directly suppress CD4 T-lymphocyte activation and proliferation. Immunobiology 2013; 218:1034-40. [PMID: 23434301 DOI: 10.1016/j.imbio.2013.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Revised: 01/01/2013] [Accepted: 01/02/2013] [Indexed: 10/27/2022]
Abstract
CD4 T cells are important regulators of the immune system and are vital for mounting a strong immune response against viral infections. Human cytomegalovirus (HCMV) is known to be a strong modulator of the innate as well as adaptive immune responses. In this study, we found that HCMV directly inhibited proliferation of CD4 T cells and rendered them unresponsive to immunological stimuli. This effect was not observed when CD4 T cells were treated with herpes simplex virus-1/2 or measles virus. When stimulated with phytohemagglutinin, concanavalin A, or phorbol myristate acetate, HCMV-treated T cells were unable to proliferate, revealing an ability of HCMV to inhibit CD4 T cell response. Furthermore, HCMV also prevented proliferation of leukemic T-cell lines. HCMV-treated CD4 T cells expressed the activation markers CD45RO and CD69, were not apoptotic and produced decreased levels of the cytokines IL-4, IFN-γ and TNF-α, compared to untreated controls. The inhibitory effect of HCMV on CD4 T cell proliferation was not mediated by HCMV gH, gB or other immunogenic glycoproteins, since intravenous immunoglobulins or gB- or gH-specific neutralizing antibodies did not prevent the suppression of T-cell proliferation. Our observations show that HCMV inhibits CD4 T cell function with potential clinical consequences for both humoral and cell-mediated immune responses.
Collapse
Affiliation(s)
- Olesja Fornara
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, 17176 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|