1
|
Einwächter H, Heiseke A, Schlitzer A, Gasteiger G, Adler H, Voehringer D, Manz MG, Ruzsics Z, Dölken L, Koszinowski UH, Sparwasser T, Reindl W, Jordan S. The Innate Immune Response to Infection Induces Erythropoietin-Dependent Replenishment of the Dendritic Cell Compartment. Front Immunol 2020; 11:1627. [PMID: 32849551 PMCID: PMC7411349 DOI: 10.3389/fimmu.2020.01627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/17/2020] [Indexed: 12/29/2022] Open
Abstract
Dendritic cells (DC) play a key role in the adaptive immune response due to their ability to present antigens and stimulate naïve T cells. Many bacteria and viruses can efficiently target DC, resulting in impairment of their immunostimulatory function or elimination. Hence, the DC compartment requires replenishment following infection to ensure continued operational readiness of the adaptive immune system. Here, we investigated the molecular and cellular mechanisms of inflammation-induced DC generation. We found that infection with viral and bacterial pathogens as well as Toll-like receptor 9 (TLR9) ligation with CpG-oligodeoxynucleotide (CpG-ODN) expanded an erythropoietin (EPO)-dependent TER119+CD11a+ cell population in the spleen that had the capacity to differentiate into TER119+CD11chigh and TER119-CD11chigh cells both in vitro and in vivo. TER119+CD11chigh cells contributed to the conventional DC pool in the spleen and specifically increased in lymph nodes draining the site of local inflammation. Our results reveal a so far undescribed inflammatory EPO-dependent pathway of DC differentiation and establish a mechanistic link between innate immune recognition of potential immunosuppressive pathogens and the maintenance of the DC pool during and after infection.
Collapse
Affiliation(s)
- Henrik Einwächter
- II. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Alexander Heiseke
- II. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Georg Gasteiger
- Institute of Systems Immunology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Heiko Adler
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Munich, Germany.,German Center of Lung Research (DZL), Giessen, Germany
| | - David Voehringer
- Department of Infection Biology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Nürnberg, Erlangen, Germany
| | - Markus G Manz
- Division of Hematology, Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Zsolt Ruzsics
- Institute of Virology, University Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Lars Dölken
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Ulrich H Koszinowski
- Max von Pettenkofer-Institute, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medicine Mainz, Johannes-Gutenberg-University Mainz, Mainz, Germany
| | - Wolfgang Reindl
- II. Medizinische Klinik, Medizinische Fakultät Mannheim, Universität Heidelberg, Mannheim, Germany
| | - Stefan Jordan
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| |
Collapse
|
2
|
Strazic Geljic I, Kucan Brlic P, Angulo G, Brizic I, Lisnic B, Jenus T, Juranic Lisnic V, Pietri GP, Engel P, Kaynan N, Zeleznjak J, Schu P, Mandelboim O, Krmpotic A, Angulo A, Jonjic S, Lenac Rovis T. Cytomegalovirus protein m154 perturbs the adaptor protein-1 compartment mediating broad-spectrum immune evasion. eLife 2020; 9:50803. [PMID: 31928630 PMCID: PMC6957316 DOI: 10.7554/elife.50803] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/03/2020] [Indexed: 12/21/2022] Open
Abstract
Cytomegaloviruses (CMVs) are ubiquitous pathogens known to employ numerous immunoevasive strategies that significantly impair the ability of the immune system to eliminate the infected cells. Here, we report that the single mouse CMV (MCMV) protein, m154, downregulates multiple surface molecules involved in the activation and costimulation of the immune cells. We demonstrate that m154 uses its cytoplasmic tail motif, DD, to interfere with the adaptor protein-1 (AP-1) complex, implicated in intracellular protein sorting and packaging. As a consequence of the perturbed AP-1 sorting, m154 promotes lysosomal degradation of several proteins involved in T cell costimulation, thus impairing virus-specific CD8+ T cell response and virus control in vivo. Additionally, we show that HCMV infection similarly interferes with the AP-1 complex. Altogether, we identify the robust mechanism employed by single viral immunomodulatory protein targeting a broad spectrum of cell surface molecules involved in the antiviral immune response.
Collapse
Affiliation(s)
- Ivana Strazic Geljic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Paola Kucan Brlic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Guillem Angulo
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Ilija Brizic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Berislav Lisnic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Tina Jenus
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Vanda Juranic Lisnic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Gian Pietro Pietri
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Pablo Engel
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Noa Kaynan
- The Lautenberg Center for General and Tumor Immunology, The BioMedical Research Institute, Hadassah Medical School, The Hebrew University, Jerusalem, Israel
| | - Jelena Zeleznjak
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Peter Schu
- Zentrum für Biochemie und Molekulare Zellbiologie Institut für Zellbiochemie, Georg-August-Universität Göttingen, Goettingen, Germany
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, The BioMedical Research Institute, Hadassah Medical School, The Hebrew University, Jerusalem, Israel
| | - Astrid Krmpotic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Ana Angulo
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Stipan Jonjic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Tihana Lenac Rovis
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
3
|
Activated dendritic cells and monocytes in HIV immunological nonresponders: HIV-induced interferon-inducible protein-10 correlates with low future CD4+ recovery. AIDS 2019; 33:1117-1129. [PMID: 30789356 PMCID: PMC6511429 DOI: 10.1097/qad.0000000000002173] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Supplemental Digital Content is available in the text Objective: To explore monocyte and dendritic cell immune responses, and their association with future CD4+ gain in treated HIV patients with suboptimal CD4+ recovery. Design: A cross-sectional study of HIV-infected, virally suppressed individuals on antiretroviral therapy for at least 24 months; 41 immunological nonresponders (INRs) (CD4+ cell count <400 cells/μl) and 26 immunological responders (CD4+ cell count >600 cells/μl). Ten HIV-infected antiretroviral therapy-naive and 10 HIV-negative healthy persons served as controls. CD4+ cell counts were registered after median 2.4 and 4.7 years. Methods: Monocyte, dendritic-cell and T-cell activation and regulatory T cells (Tregs) were analyzed by flow cytometry. In INR and immunological responder subgroups matched on age and nadir CD4+ cell count, upregulation of interferon-inducible protein-10 (IP-10) and indoleamine 2,3-dioxygenase in monocytes and dendritic cells and cytokines in cell supernatants were measured in vitro in peripheral blood mononuclear cells stimulated with aldrithiol-2-inactivated HIV-1. Results: The INR group displayed higher spontaneous activation of both monocytes (HLA-DR+) and myeloid and plasmacytoid dendritic cells (HLA-DR+, CD83+ and CD86+) compared with immunological responders, and this was associated with increased T-cell activation (CD38+HLA-DR+), an effector memory T-cell phenotype and activated Tregs. The IP-10 response in monocytes after in-vitro HIV stimulation was negatively associated with prospective CD4+ gain. IP-10, indoleamine 2,3-dioxygenase and cytokines levels were comparable between the groups, but inversely correlated with activated Tregs in INRs. Conclusion: HIV-infected individuals with suboptimal immune recovery demonstrated more activated monocytes and in particular dendritic cells, compared with patients with acceptable CD4+ gain. A high level of HIV-specific IP-10 expression in monocytes may be predictive of future CD4+ recovery.
Collapse
|
4
|
El-Mokhtar MA, Bauer A, Madela J, Voigt S. Cellular distribution of CD200 receptor in rats and its interaction with cytomegalovirus e127 protein. Med Microbiol Immunol 2018; 207:307-318. [PMID: 30032349 DOI: 10.1007/s00430-018-0552-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 07/17/2018] [Indexed: 12/11/2022]
Abstract
CD200 is a membrane protein that interacts with CD200R on the surface of immune cells and delivers an inhibitory signal. In this study, we characterized the distribution of inhibitory CD200R in rats. In addition, we investigated if e127, a homologue of rat CD200 expressed by rat cytomegalovirus (RCMV), can suppress immune functions in vitro. RT-PCR analysis was carried out to test the expression of CD200R in different rat tissues and flow cytometry was performed to characterize CD200R at the cellular level. To test the inhibitory functions of e127, a co-culture system was utilized in which immune cells were incubated with e127-expressing cells. The strongest CD200R expression was detected in lymphoid organs such as bone marrow and spleen. Flow cytometry analyses showed that CD200R+ cells were mainly CD4- dendritic cells (DC) and CD4+ T cells in the spleen. In blood, nearly all monocytes and granulocytes expressed CD200R and in bone marrow the NKRP1low subset of natural killer cells highly expressed CD200R. In addition, both peritoneal macrophages and the NR8383 macrophage cell line carried CD200R. At the functional level, viral e127 conferred an inhibitory signal on TNFα and IL6 cytokine release from IFNγ-stimulated macrophages. However, e127 did not affect the cytotoxic activity of DC. CD200R in the rat is mainly expressed on myeloid cells but also on non-myeloid cell subsets, and RCMV e127 can deliver inhibitory signals to immune cells by engaging CD200R. The RCMV model provides a useful tool to study potential immune evasion mechanisms of the herpesviridae and opens new avenues for understanding and controlling herpesvirus infections.
Collapse
Affiliation(s)
- Mohamed A El-Mokhtar
- Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany.,Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Agnieszka Bauer
- Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany
| | - Julia Madela
- Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany
| | - Sebastian Voigt
- Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany. .,Department of Pediatric Oncology/Hematology/SCT, Charité-Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
5
|
Korbecki J, Gutowska I, Kojder I, Jeżewski D, Goschorska M, Łukomska A, Lubkowska A, Chlubek D, Baranowska-Bosiacka I. New extracellular factors in glioblastoma multiforme development: neurotensin, growth differentiation factor-15, sphingosine-1-phosphate and cytomegalovirus infection. Oncotarget 2018; 9:7219-7270. [PMID: 29467963 PMCID: PMC5805549 DOI: 10.18632/oncotarget.24102] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 01/02/2018] [Indexed: 11/25/2022] Open
Abstract
Recent years have seen considerable progress in understanding the biochemistry of cancer. For example, more significance is now assigned to the tumor microenvironment, especially with regard to intercellular signaling in the tumor niche which depends on many factors secreted by tumor cells. In addition, great progress has been made in understanding the influence of factors such as neurotensin, growth differentiation factor-15 (GDF-15), sphingosine-1-phosphate (S1P), and infection with cytomegalovirus (CMV) on the 'hallmarks of cancer' in glioblastoma multiforme. Therefore, in the present work we describe the influence of these factors on the proliferation and apoptosis of neoplastic cells, cancer stem cells, angiogenesis, migration and invasion, and cancer immune evasion in a glioblastoma multiforme tumor. In particular, we discuss the effect of neurotensin, GDF-15, S1P (including the drug FTY720), and infection with CMV on tumor-associated macrophages (TAM), microglial cells, neutrophil and regulatory T cells (Treg), on the tumor microenvironment. In order to better understand the role of the aforementioned factors in tumoral processes, we outline the latest models of intratumoral heterogeneity in glioblastoma multiforme. Based on the most recent reports, we discuss the problems of multi-drug therapy in treating glioblastoma multiforme.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland.,Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biała, 43-309 Bielsko-Biała, Poland
| | - Izabela Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Ireneusz Kojder
- Department of Applied Neurocognitivistics, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland.,Department of Neurosurgery, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Dariusz Jeżewski
- Department of Applied Neurocognitivistics, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland.,Department of Neurosurgery, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Marta Goschorska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Agnieszka Łukomska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Anna Lubkowska
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| |
Collapse
|
6
|
Touma W, Brunstein CG, Cao Q, Miller JS, Curtsinger J, Verneris MR, Bachanova V. Dendritic Cell Recovery Impacts Outcomes after Umbilical Cord Blood and Sibling Donor Transplantation for Hematologic Malignancies. Biol Blood Marrow Transplant 2017; 23:1925-1931. [PMID: 28729150 DOI: 10.1016/j.bbmt.2017.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 07/12/2017] [Indexed: 11/18/2022]
Abstract
Dendritic cells (DCs) orchestrate immune responses after allogeneic hematopoietic cell transplantation (HCT). We studied the association of donor myeloid DCs (mDCs) and plasmacytoid DCs (pDCs) recovery in the landmark analysis of umbilical cord blood (UCB) and matched related donor (RD) HCT. Eighty patients (42 UCB and 38 RD recipients) with a day 100 blood sample were included in the analysis. Median age was 51 years (range, 20 to 71). Most patients had acute leukemia (50%) or lymphoma (23%) and received reduced-intensity conditioning (75%). After transplantation, UCB recipients had higher DC counts than RD recipients reaching normal levels at day 100 after transplantation (UCB median 4.7 cells/µL versus RD median 1.7 cells/µL). UCB recipients with high day 100 pDCs levels (≥ median) had 2-fold lower risk of relapse compared with those with pDClow (14% versus 28%, P = .29) and a trend to improved 1-year survival in multivariate analysis with hazard ratio of .22 (95% confidence interval, .04 to 1.05; P = .057). Cytomegalovirus (CMV) reactivation had adverse impact on DC reconstitution at day 100 in both UCB and RD groups and almost exclusively affected the mDC subset (CMV reactivation: mDC 3.2 cells/µL versus no CMV reactivation: 7.8 cells/µL; P = .004). Collectively, these data suggest that high levels of circulating pDCs at day 100 after UCB transplantation confer a survival advantage at 1 year.
Collapse
Affiliation(s)
- Waseem Touma
- Blood and Marrow Transplantation Program, University of Minnesota, Minneapolis, Minnesota
| | - Claudio G Brunstein
- Blood and Marrow Transplantation Program, University of Minnesota, Minneapolis, Minnesota
| | - Qing Cao
- Biostatistics and Informatics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey S Miller
- Blood and Marrow Transplantation Program, University of Minnesota, Minneapolis, Minnesota
| | - Julie Curtsinger
- Blood and Marrow Transplantation Program, University of Minnesota, Minneapolis, Minnesota
| | - Michael R Verneris
- Colorado School of Medicine, Children's Hospital Colorado, Aurora, Colorado
| | - Veronika Bachanova
- Blood and Marrow Transplantation Program, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
7
|
Karimi MH, Shariat A, Yaghobi R, Mokhtariazad T, Moazzeni SM. Role of cytomegalovirus on the maturation and function of monocyte derived dendritic cells of liver transplant patients. World J Transplant 2016; 6:336-346. [PMID: 27358779 PMCID: PMC4919738 DOI: 10.5500/wjt.v6.i2.336] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/24/2016] [Accepted: 04/22/2016] [Indexed: 02/05/2023] Open
Abstract
AIM: To study the impact of association between cytomegalovirus (CMV) pathogenesis with dendritic cell (DC) maturation and function was evaluated in CMV reactivated liver transplanted patients in comparing with non-reactivated ones, and healthy controls.
METHODS: Monocyte derived dendritic cells (MoDCs) was generated from collected ethylenediaminetetraacetic acid-treated blood samples from patient groups and controls. In these groups, expression rates and mean fluorescent intensity of DC markers were evaluated using flowcytometry technique. Secretion of cytokines including: interleukin (IL)-6, IL-12 and IL-23 were determined using enzyme-linked immunosorbent assay methods. The gene expression of toll-like receptor 2 (TLR2), TLR4 and IL-23 were analyzed using in-house real-time polymerase chain reaction protocols.
RESULTS: Results have been shown significant decreases in: Expression rates of MoDC markers including CD83, CD1a and human leukocyte antigen DR (HLA-DR), the mean fluorescence intensitys for CD1a and HLA-DR, and secretion of IL-12 in CMV reactivated compared with non-reactivated liver transplanted patients. On the other hand, significant increases have been shown in the secretions of IL-6 and IL-23 and gene expression levels of TLR2, TLR4 and IL-23 from MoDCs in CMV reactivated compared with non-reactivated liver transplanted recipients.
CONCLUSION: DC functional defects in CMV reactivated recipients, such as decrease in expression of DC maturation markers, increase in secretion of proinflammatory cytokines, and TLRs can emphasize on the importance of CMV infectivity in development of liver rejection in transplanted patients.
Collapse
|
8
|
[Immunomonitoring for cytomegalovirus infection in kidney transplantation: Development and prospects]. Nephrol Ther 2015. [PMID: 26206770 DOI: 10.1016/j.nephro.2015.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cytomegalovirus infection in kidney transplantation is associated with increased morbidity and mortality through direct and indirect effects. International guidelines had been recently updated, focusing on diagnostic, prevention strategies and curative treatment. Cytomegalovirus-specific immune response plays also an important function in controlling the virus. Here, we propose to present the different components of this specific immune response and the advantages of immune monitoring for patient's management: identification of patients who require a treatment, adaptation of curative treatment length, guidance for resistance genotypic testing.
Collapse
|
9
|
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: 1.6] [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
|
10
|
Del Prete A, Luganini A, Scutera S, Rossi S, Anselmo A, Greco D, Landolfo S, Badolato R, Gribaudo G, Sozzani S, Musso T. Interferon-α production by plasmacytoid dendritic cells is dispensable for an effective anti-cytomegalovirus response in adaptor protein-3-deficient mice. J Interferon Cytokine Res 2014; 35:232-8. [PMID: 25333950 DOI: 10.1089/jir.2013.0110] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Adaptor protein-3 (AP-3) is a heterotetrameric complex, which regulates vesicular trafficking. Mutations of the β3A subunit cause the Hermansky-Pudlak syndrome type 2 (HPS-2), a rare genetic disease characterized by albinism, platelet defects, and recurrent infections. Likewise, pearl mice, which lack functional AP-3, show several HPS-2 defects. The AP-3 absence results in defective toll-like receptor trafficking and signaling in dendritic cells (DC), but its effect on the efficiency of the in vivo antiviral response is unclear. We evaluated the impact of AP-3 deficiency on the distribution of DC subsets, interferon (IFN) production, and the susceptibility to murine cytomegalovirus (MCMV) infection. Pearl mice showed a distribution and frequency of conventional (cDC) and plasmacytoid DC (pDC) similar to that of wild-type mice both before and after MCMV infection. Moreover, pearl mice controlled MCMV infection even at high virus doses and showed a normal production of IFN-α. Since pDC, but not cDC, from pearl mice showed an impaired IFN-α and tumor necrosis factor-α production in response to prototypic DNA (MCMV and Herpes Simplex virus) or RNA (Vesicular Stomatitis virus) viruses in vitro, it is likely that MCMV infection can be controlled in vivo independently of an efficient production of IFN-α by pDC, and that the AP-3 complex has a minimal impact on protective antiviral responses.
Collapse
Affiliation(s)
- Annalisa Del Prete
- 1 Department of Molecular and Translational Medicine, University of Brescia , Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Avdic S, McSharry BP, Slobedman B. Modulation of dendritic cell functions by viral IL-10 encoded by human cytomegalovirus. Front Microbiol 2014; 5:337. [PMID: 25071749 PMCID: PMC4081832 DOI: 10.3389/fmicb.2014.00337] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/17/2014] [Indexed: 12/22/2022] Open
Abstract
Human cytomegalovirus (HCMV), a clinically important β-herpesvirus, is a master of evasion and modulation of the host immune system, including inhibition of a number of dendritic cell (DC) functions. DCs play a central role in co-ordination of the immune response against pathogens and any disturbance of DCs functions can result in a cascade effect on a range of immune cells. Recently, the HCMV gene UL111A, which encodes viral homologs of human interleukin 10, has been identified as a strong suppressor of a number of DCs functions. In this mini review, we focus on HCMV-encoded viral IL-10-mediated inhibitory effects on DCs and implications for the development of an effective HCMV vaccine.
Collapse
Affiliation(s)
- Selmir Avdic
- Human Cytomegalovirus Research Group, Discipline of Infectious Diseases and Immunology, University of Sydney Camperdown, NSW, Australia
| | - Brian P McSharry
- Human Cytomegalovirus Research Group, Discipline of Infectious Diseases and Immunology, University of Sydney Camperdown, NSW, Australia
| | - Barry Slobedman
- Human Cytomegalovirus Research Group, Discipline of Infectious Diseases and Immunology, University of Sydney Camperdown, NSW, Australia ; Centre for Virus Research, Westmead Millennium Institute Westmead, NSW, Australia
| |
Collapse
|
12
|
Glover TE, Kew VG, Reeves MB. Rapamycin does not inhibit human cytomegalovirus reactivation from dendritic cells in vitro. J Gen Virol 2014; 95:2260-2266. [PMID: 24986086 PMCID: PMC4165932 DOI: 10.1099/vir.0.066332-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection and reactivation are a major cause of morbidity in immune-suppressed patients. Interestingly, epidemiological studies have shown that patients administered the mammalian target of rapamycin (mTOR) inhibitor, sirolimus (rapamycin), exhibit more favourable outcomes, suggestive of activity against HCMV in vivo. Given its relative lack of activity against lytic infection, it is postulated that rapamycin inhibits HCMV reactivation. Here, we showed that rapamycin administered acutely or chronically has little impact on induction of immediate early (IE) gene expression in experimentally latent dendritic cells or cells from naturally latent individuals. Furthermore, we extended these observations to include other inhibitors of mTORC1 and mTORC 2, which similarly have minimal effects on induction of IE gene expression from latency. Taken together, these data suggest that favourable outcomes associated with sirolimus are attributable to indirect effects that influence HCMV reactivation, rather than a direct mechanistic action against HCMV itself.
Collapse
Affiliation(s)
- Thomas E Glover
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK
| | - Verity G Kew
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK
| | - Matthew B Reeves
- Institute of Immunity & Transplantation, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, UK.,Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
13
|
Hertel L. Human cytomegalovirus tropism for mucosal myeloid dendritic cells. Rev Med Virol 2014; 24:379-95. [PMID: 24888709 DOI: 10.1002/rmv.1797] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/01/2014] [Accepted: 05/07/2014] [Indexed: 02/01/2023]
Abstract
Human CMV infections are a serious source of morbidity and mortality for immunocompromised patients and for the developing fetus. Because of this, the development of new strategies to prevent CMV acquisition and transmission is a top priority. Myeloid dendritic cells (DC) residing in the oral and nasal mucosae are among the first immune cells to encounter CMV during entry and greatly contribute to virus dissemination, reactivation from latency, and horizontal spread. Albeit affected by the immunoevasive tactics of CMV, mucosal DC remain potent inducers of cellular and humoral immune responses against this virus. Their natural functions could thus be exploited to generate long-lasting protective immunity against CMV by vaccination via the oronasal mucosae. Although related, epithelial Langerhans-type DC and dermal monocyte-derived DC interact with CMV in dramatically different ways. Whereas immature monocyte-derived DC are fully permissive to infection, for instance, immature Langerhans-type DC are completely resistant. Understanding these differences is essential to design innovative vaccines and new antiviral compounds to protect these cells from CMV infection in vivo.
Collapse
Affiliation(s)
- Laura Hertel
- Center for Immunobiology and Vaccine Development, Children's Hospital Oakland Research Institute, Oakland, CA, 94609, USA
| |
Collapse
|
14
|
Cytomegalovirus infections of the adult human nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2014; 123:307-18. [DOI: 10.1016/b978-0-444-53488-0.00014-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Human cytomegalovirus infection of langerhans-type dendritic cells does not require the presence of the gH/gL/UL128-131A complex and is blocked after nuclear deposition of viral genomes in immature cells. J Virol 2013; 88:403-16. [PMID: 24155395 DOI: 10.1128/jvi.03062-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human cytomegalovirus (CMV) enters its host via the oral and genital mucosae. Langerhans-type dendritic cells (LC) are the most abundant innate immune cells at these sites, where they constitute a first line of defense against a variety of pathogens. We previously showed that immature LC (iLC) are remarkably resistant to CMV infection, while mature LC (mLC) are more permissive, particularly when exposed to clinical-strain-like strains of CMV, which display a pentameric complex consisting of the viral glycoproteins gH, gL, UL128, UL130, and UL131A on their envelope. This complex was recently shown to be required for the infection of immature monocyte-derived dendritic cells. We thus sought to establish if the presence of this complex is also necessary for virion penetration of LC and if defects in entry might be the source of iLC resistance to CMV. Here we report that the efficiency of LC infection is reduced, but not completely abolished, in the absence of the pentameric complex. While virion penetration and nuclear deposition of viral genomes are not impaired in iLC, the transcription of the viral immediate early genes UL122 and UL123 and of the delayed early gene UL50 is substantially lower than that in mLC. Together, these data show that the UL128, UL130, and UL131A proteins are dispensable for CMV entry into LC and that progression of the viral cycle in iLC is restricted at the step of viral gene expression.
Collapse
|
16
|
Walton S, Mandaric S, Oxenius A. CD4 T cell responses in latent and chronic viral infections. Front Immunol 2013; 4:105. [PMID: 23717308 PMCID: PMC3651995 DOI: 10.3389/fimmu.2013.00105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/22/2013] [Indexed: 12/24/2022] Open
Abstract
The spectrum of tasks which is fulfilled by CD4 T cells in the setting of viral infections is large, ranging from support of CD8 T cells and humoral immunity to exertion of direct antiviral effector functions. While our knowledge about the differentiation pathways, plasticity, and memory of CD4 T cell responses upon acute infections or immunizations has significantly increased during the past years, much less is still known about CD4 T cell differentiation and their beneficial or pathological functions during persistent viral infections. In this review we summarize current knowledge about the differentiation, direct or indirect antiviral effector functions, and the regulation of virus-specific CD4 T cells in the setting of persistent latent or active chronic viral infections with a particular emphasis on herpes virus infections for the former and chronic lymphocytic choriomeningitis virus infection for the latter.
Collapse
Affiliation(s)
- Senta Walton
- Department of Microbiology and Immunology, School of Pathology and Laboratory Medicine, University of Western Australia Nedlands, WA, Australia
| | | | | |
Collapse
|
17
|
Noriega V, Redmann V, Gardner T, Tortorella D. Diverse immune evasion strategies by human cytomegalovirus. Immunol Res 2013; 54:140-51. [PMID: 22454101 DOI: 10.1007/s12026-012-8304-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Members of the Herpesviridae family have the capacity to undergo both lytic and latent infection to establish a lifelong relationship with their host. Following primary infection, human cytomegalovirus (HCMV) can persist as a subclinical, recurrent infection for the lifetime of an individual. This quiescent portion of its life cycle is termed latency and is associated with periodic bouts of reactivation during times of immunosuppression, inflammation, or stress. In order to exist indefinitely and establish infection, HCMV encodes a multitude of immune modulatory mechanisms devoted to escaping the host antiviral response. HCMV has become a paradigm for studies of viral immune evasion of antigen presentation by both major histocompatibility complex (MHC) class I and II molecules. By restricting the presentation of viral antigens during both productive and latent infection, HCMV limits elimination by the human immune system. This review will focus on understanding how the virus manipulates the pathways of antigen presentation in order to modulate the host response to infection.
Collapse
Affiliation(s)
- Vanessa Noriega
- Department of Microbiology, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA
| | | | | | | |
Collapse
|
18
|
Cytomegalovirus (CMV)-dependent and -independent changes in the aging of the human immune system: A transcriptomic analysis. Exp Gerontol 2013; 48:305-12. [DOI: 10.1016/j.exger.2012.12.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 12/06/2012] [Accepted: 12/26/2012] [Indexed: 11/17/2022]
|
19
|
Busche A, Jirmo AC, Welten SPM, Zischke J, Noack J, Constabel H, Gatzke AK, Keyser KA, Arens R, Behrens GMN, Messerle M. Priming of CD8+ T cells against cytomegalovirus-encoded antigens is dominated by cross-presentation. THE JOURNAL OF IMMUNOLOGY 2013; 190:2767-77. [PMID: 23390296 DOI: 10.4049/jimmunol.1200966] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
CMV can infect dendritic cells (DCs), and direct Ag presentation could, therefore, lead to the priming of CMV-specific CD8(+) T cells. However, CMV-encoded immune evasins severely impair Ag presentation in the MHC class I pathway; thus, it is widely assumed that cross-presentation drives the priming of antiviral T cells. We assessed the contribution of direct versus cross priming in mouse CMV (MCMV) infection using recombinant viruses. DCs infected with an MCMV strain encoding the gB498 epitope from HSV-1 were unable to stimulate in vitro naive gB498-specific CD8(+) T cells from TCR transgenic mice. Infection of C57BL/6 mice with this recombinant virus led, however, to the generation of abundant numbers of gB498-specific T cells in vivo. Of the DC subsets isolated from infected mice, only CD8α(+) DCs were able to stimulate naive T cells, suggesting that this DC subset cross-presents MCMV-encoded Ag in vivo. Upon infection of mice with MCMV mutants encoding Ag that can either be well or hardly cross-presented, mainly CD8(+) T cells specific for cross-presented epitopes were generated. Moreover, even in the absence of immune evasion genes interfering with MHC class I-mediated Ag presentation, priming of T cells to Ag that can only be presented directly was not observed. We conclude that the host uses mainly DCs capable of cross-presentation to induce the CMV-specific CD8(+) T cell response during primary, acute infection and discuss the implications for the development of a CMV vaccine.
Collapse
Affiliation(s)
- Andreas Busche
- Department of Virology, Hannover Medical School, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Arpaia N, Barton GM. Toll-like receptors: key players in antiviral immunity. Curr Opin Virol 2011; 1:447-54. [PMID: 22440908 DOI: 10.1016/j.coviro.2011.10.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 10/08/2011] [Indexed: 12/24/2022]
Abstract
TLRs are a family of innate receptors whose specificities are predetermined in the germline. Therefore, TLRs have evolved to recognize conserved features of microbes. Viruses typically lack the conserved features common to other pathogen classes, so the innate immune system has evolved to recognize viral nucleic acid as a hallmark of viral infection. In this review we discuss examples of TLR-mediated viral recognition and the functional consequences of this recognition for antiviral immunity.
Collapse
Affiliation(s)
- Nicholas Arpaia
- Division of Immunology & Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | | |
Collapse
|
21
|
Activation of the human immune system by chemotherapeutic or targeted agents combined with the oncolytic parvovirus H-1. BMC Cancer 2011; 11:464. [PMID: 22029859 PMCID: PMC3234202 DOI: 10.1186/1471-2407-11-464] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 10/26/2011] [Indexed: 01/08/2023] Open
Abstract
Background Parvovirus H-1 (H-1PV) infects and lyses human tumor cells including melanoma, hepatoma, gastric, colorectal, cervix and pancreatic cancers. We assessed whether the beneficial effects of chemotherapeutic agents or targeted agents could be combined with the oncolytic and immunostimmulatory properties of H-1PV. Methods Using human ex vivo models we evaluated the biological and immunological effects of H-1PV-induced tumor cell lysis alone or in combination with chemotherapeutic or targeted agents in human melanoma cells +/- characterized human cytotoxic T-cells (CTL) and HLA-A2-restricted dendritic cells (DC). Results H-1PV-infected MZ7-Mel cells showed a clear reduction in cell viability of >50%, which appeared to occur primarily through apoptosis. This correlated with viral NS1 expression levels and was enhanced by combination with chemotherapeutic agents or sunitinib. Tumor cell preparations were phagocytosed by DC whose maturation was measured according to the treatment administered. Immature DC incubated with H-1PV-induced MZ7-Mel lysates significantly increased DC maturation compared with non-infected or necrotic MZ7-Mel cells. Tumor necrosis factor-α and interleukin-6 release was clearly increased by DC incubated with H-1PV-induced SK29-Mel tumor cell lysates (TCL) and was also high with DC-CTL co-cultures incubated with H-1PV-induced TCL. Similarly, DC co-cultures with TCL incubated with H-1PV combined with cytotoxic agents or sunitinib enhanced DC maturation to a greater extent than cytotoxic agents or sunitinib alone. Again, these combinations increased pro-inflammatory responses in DC-CTL co-cultures compared with chemotherapy or sunitinib alone. Conclusions In our human models, chemotherapeutic or targeted agents did not only interfere with the pronounced immunomodulatory properties of H-1PV, but also reinforced drug-induced tumor cell killing. H-1PV combined with cisplatin, vincristine or sunitinib induced effective immunostimulation via a pronounced DC maturation, better cytokine release and cytotoxic T-cell activation compared with agents alone. Thus, the clinical assessment of H-1PV oncolytic tumor therapy not only alone but also in combination strategies is warranted.
Collapse
|
22
|
Herpesviruses and intermediate filaments: close encounters with the third type. Viruses 2011; 3:1015-40. [PMID: 21994768 PMCID: PMC3185793 DOI: 10.3390/v3071015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 06/07/2011] [Accepted: 06/24/2011] [Indexed: 01/29/2023] Open
Abstract
Intermediate filaments (IF) are essential to maintain cellular and nuclear integrity and shape, to manage organelle distribution and motility, to control the trafficking and pH of intracellular vesicles, to prevent stress-induced cell death, and to support the correct distribution of specific proteins. Because of this, IF are likely to be targeted by a variety of pathogens, and may act in favor or against infection progress. As many IF functions remain to be identified, however, little is currently known about these interactions. Herpesviruses can infect a wide variety of cell types, and are thus bound to encounter the different types of IF expressed in each tissue. The analysis of these interrelationships can yield precious insights into how IF proteins work, and into how viruses have evolved to exploit these functions. These interactions, either known or potential, will be the focus of this review.
Collapse
|
23
|
Cook CH, Trgovcich J. Cytomegalovirus reactivation in critically ill immunocompetent hosts: a decade of progress and remaining challenges. Antiviral Res 2011; 90:151-9. [PMID: 21439328 PMCID: PMC3129598 DOI: 10.1016/j.antiviral.2011.03.179] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Revised: 03/14/2011] [Accepted: 03/15/2011] [Indexed: 01/05/2023]
Abstract
Human cytomegalovirus (HCMV) is an undisputed pathogen in humans with severe immune compromise, which has historically been thought to carry little consequence in immunocompetent hosts. During the past decade, however, accumulating data suggest that significant numbers of immunocompetent humans reactivate HCMV during critical illness, and that these reactivation episodes are associated with worsened outcomes. Because most people are infected with this ubiquitous virus by adulthood, confirming pathogenicity has now become a clinical priority. In this article, we will review the incidence and implications of reactivation, the relevant immune responses and reactivation triggers relevant to the immunocompetent host. We will summarize the progress made during the past ten years, outline the work ongoing in this field, and identify the major gaps remaining in our emerging understanding of this phenomenon.
Collapse
Affiliation(s)
- Charles H Cook
- Department of Surgery, The Ohio State University, Columbus, OH 43210, USA.
| | | |
Collapse
|
24
|
Lugo-Villarino G, Neyrolles O. Editorial: How to play tag? DC-SIGN shows the way! J Leukoc Biol 2011; 89:321-3. [PMID: 21357245 DOI: 10.1189/jlb.1010547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
25
|
Slavuljica I, Busche A, Babić M, Mitrović M, Gašparović I, Cekinović D, Markova Car E, Pernjak Pugel E, Ciković A, Lisnić VJ, Britt WJ, Koszinowski U, Messerle M, Krmpotić A, Jonjić S. Recombinant mouse cytomegalovirus expressing a ligand for the NKG2D receptor is attenuated and has improved vaccine properties. J Clin Invest 2010; 120:4532-45. [PMID: 21099111 DOI: 10.1172/jci43961] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Accepted: 09/22/2010] [Indexed: 12/21/2022] Open
Abstract
Human CMV (HCMV) is a major cause of morbidity and mortality in both congenitally infected and immunocompromised individuals. Development of an effective HCMV vaccine would help protect these vulnerable groups. NK group 2, member D (NKG2D) is a potent activating receptor expressed by cells of the innate and adaptive immune systems. Its importance in HCMV immune surveillance is indicated by the elaborative evasion mechanisms evolved by the virus to avoid NKG2D. In order to study this signaling pathway, we engineered a recombinant mouse CMV expressing the high-affinity NKG2D ligand RAE-1γ (RAE-1γMCMV). Expression of RAE-1γ by MCMV resulted in profound virus attenuation in vivo and lower latent viral DNA loads. RAE-1γMCMV infection was efficiently controlled by immunodeficient hosts, including mice lacking type I interferon receptors or immunosuppressed by sublethal γ-irradiation. Features of MCMV infection in neonates were also diminished. Despite tight innate immune control, RAE-1γMCMV infection elicited strong and long-lasting protective immunity. Maternal RAE-1γMCMV immunization protected neonatal mice from MCMV disease via placental transfer of antiviral Abs. Despite strong selective pressure, the RAE-1γ transgene did not exhibit sequence variation following infection. Together, our results indicate that use of a recombinant virus encoding the ligand for an activating NK cell receptor could be a powerful approach to developing a safe and immunogenic HCMV vaccine.
Collapse
Affiliation(s)
- Irena Slavuljica
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Loewendorf A, Benedict CA. Modulation of host innate and adaptive immune defenses by cytomegalovirus: timing is everything. J Intern Med 2010; 267:483-501. [PMID: 20433576 PMCID: PMC2902254 DOI: 10.1111/j.1365-2796.2010.02220.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Human cytomegalovirus (HCMV) (HHV-5, a beta-herpesvirus) causes the vast majority of infection-related congenital birth defects, and can trigger severe disease in immune suppressed individuals. The high prevalence of societal infection, the establishment of lifelong persistence and the growing number of immune-related diseases where HCMV is touted as a potential promoter is slowly heightening public awareness to this virus. The millions of years of co-evolution between CMV and the immune system of its host provides for a unique opportunity to study immune defense strategies, and pathogen counterstrategies. Dissecting the timing of the cellular and molecular processes that regulate innate and adaptive immunity to this persistent virus has revealed a complex defense network that is shaped by CMV immune modulation, resulting in a finely tuned host-pathogen relationship.
Collapse
Affiliation(s)
- A Loewendorf
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | | |
Collapse
|
27
|
Baranek T, Zucchini N, Dalod M. Plasmacytoid dendritic cells and the control of herpesvirus infections. Viruses 2009; 1:383-419. [PMID: 21994554 PMCID: PMC3185500 DOI: 10.3390/v1030383] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 10/01/2009] [Accepted: 10/08/2009] [Indexed: 01/04/2023] Open
Abstract
Type-I interferons (IFN-I) are cytokines essential for vertebrate antiviral defense, including against herpesviruses. IFN-I have potent direct antiviral activities and also mediate a multiplicity of immunoregulatory functions, which can either promote or dampen antiviral adaptive immune responses. Plasmacytoid dendritic cells (pDCs) are the professional producers of IFN-I in response to many viruses, including all of the herpesviruses tested. There is strong evidence that pDCs could play a major role in the initial orchestration of both innate and adaptive antiviral immune responses. Depending on their activation pattern, pDC responses may be either protective or detrimental to the host. Here, we summarize and discuss current knowledge regarding pDC implication in the physiopathology of mouse and human herpesvirus infections, and we discuss how pDC functions could be manipulated in immunotherapeutic settings to promote health over disease.
Collapse
Affiliation(s)
- Thomas Baranek
- Université de la Méditerranée, Centre d’Immunologie de Marseille-Luminy, Parc Scientifique & Technologique de Luminy, Case 906, F13288 Marseille, Cedex 09, France; E-Mails: (T.B.); (N.Z.)
- Institut National de la Santé et de la Recherche Médicale (INSERM), U631, Centre d’Immunologie de Marseille-Luminy, Parc Scientifique & Technologique de Luminy, Case 906, F13288 Marseille, Cedex 09, France
- Centre National de la Recherche Scientifique (CNRS), UMR6102, Centre d’Immunologie de Marseille-Luminy, Parc Scientifique & Technologique de Luminy, Case 906, F13288 Marseille, Cedex 09, France
| | - Nicolas Zucchini
- Université de la Méditerranée, Centre d’Immunologie de Marseille-Luminy, Parc Scientifique & Technologique de Luminy, Case 906, F13288 Marseille, Cedex 09, France; E-Mails: (T.B.); (N.Z.)
- Institut National de la Santé et de la Recherche Médicale (INSERM), U631, Centre d’Immunologie de Marseille-Luminy, Parc Scientifique & Technologique de Luminy, Case 906, F13288 Marseille, Cedex 09, France
- Centre National de la Recherche Scientifique (CNRS), UMR6102, Centre d’Immunologie de Marseille-Luminy, Parc Scientifique & Technologique de Luminy, Case 906, F13288 Marseille, Cedex 09, France
| | - Marc Dalod
- Université de la Méditerranée, Centre d’Immunologie de Marseille-Luminy, Parc Scientifique & Technologique de Luminy, Case 906, F13288 Marseille, Cedex 09, France; E-Mails: (T.B.); (N.Z.)
- Institut National de la Santé et de la Recherche Médicale (INSERM), U631, Centre d’Immunologie de Marseille-Luminy, Parc Scientifique & Technologique de Luminy, Case 906, F13288 Marseille, Cedex 09, France
- Centre National de la Recherche Scientifique (CNRS), UMR6102, Centre d’Immunologie de Marseille-Luminy, Parc Scientifique & Technologique de Luminy, Case 906, F13288 Marseille, Cedex 09, France
| |
Collapse
|