1
|
Abstract
Heatstroke, which is associated with circulatory failure and multiple organ dysfunction, is a heat stress-induced life-threatening condition characterized by a raised core body temperature and central nervous system dysfunction. As global warming continues to worsen, heatstroke is expected to become the leading cause of death globally. Despite the severity of this condition, the detailed mechanisms that underlie the pathogenesis of heatstroke still remain largely unknown. Z-DNA-binding protein 1 (ZBP1), also referred to as DNA-dependent activator of IFN-regulatory factors (DAI) and DLM-1, was initially identified as a tumor-associated and interferon (IFN)-inducible protein, but has recently been reported to be a Z-nucleic acid sensor that regulates cell death and inflammation; however, its biological function is not yet fully understood. In the present study, a brief review of the main regulators is presented, in which the Z-nucleic acid sensor ZBP1 was identified to be a significant factor in regulating the pathological characteristics of heatstroke through ZBP1-dependent signaling. Thus, the lethal mechanism of heatstroke is revealed, in addition to a second function of ZBP1 other than as a nucleic acid sensor.
Collapse
Affiliation(s)
- Fanglin Li
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Jiayi Deng
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiuli He
- Department of Nephrology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yanjun Zhong
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
2
|
Holtappels R, Podlech J, Freitag K, Lemmermann NA, Reddehase MJ. Memory CD8 T Cells Protect against Cytomegalovirus Disease by Formation of Nodular Inflammatory Foci Preventing Intra-Tissue Virus Spread. Viruses 2022; 14:v14061145. [PMID: 35746617 PMCID: PMC9229300 DOI: 10.3390/v14061145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 12/05/2022] Open
Abstract
Cytomegaloviruses (CMVs) are controlled by innate and adaptive immune responses in an immunocompetent host while causing multiple organ diseases in an immunocompromised host. A risk group of high clinical relevance comprises transiently immunocompromised recipients of hematopoietic cell transplantation (HCT) in the “window of risk” between eradicative therapy of hematopoietic malignancies and complete reconstitution of the immune system. Cellular immunotherapy by adoptive transfer of CMV-specific CD8 T cells is an option to prevent CMV disease by controlling a primary or reactivated infection. While experimental models have revealed a viral epitope-specific antiviral function of cognate CD8 T cells, the site at which control is exerted remained unidentified. The observation that remarkably few transferred cells protect all organs may indicate an early blockade of virus dissemination from a primary site of productive infection to various target organs. Alternatively, it could indicate clonal expansion of a few transferred CD8 T cells for preventing intra-tissue virus spread after successful initial organ colonization. Our data in the mouse model of murine CMV infection provide evidence in support of the second hypothesis. We show that transferred cells vigorously proliferate to prevent virus spread, and thus viral histopathology, by confining and eventually resolving tissue infection within nodular inflammatory foci.
Collapse
|
3
|
Xu J, Liu X, Zhang X, Marshall B, Dong Z, Smith SB, Espinosa-Heidmann DG, Zhang M. Retinal and Choroidal Pathologies in Aged BALB/c Mice Following Systemic Neonatal Murine Cytomegalovirus Infection. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1787-1804. [PMID: 34197777 PMCID: PMC8485058 DOI: 10.1016/j.ajpath.2021.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 11/30/2022]
Abstract
Although pathologies associated with acute virus infections have been extensively studied, the effects of long-term latent virus infections are less well understood. Human cytomegalovirus, which infects 50% to 80% of humans, is usually acquired during early life and persists in a latent state for the lifetime. The purpose of this study was to determine whether systemic murine cytomegalovirus (MCMV) infection acquired early in life disseminates to and becomes latent in the eye and if ocular MCMV can trigger in situ inflammation and occurrence of ocular pathology. This study found that neonatal infection of BALB/c mice with MCMV resulted in dissemination of virus to the eye, where it localized principally to choroidal endothelia and pericytes and less frequently to the retinal pigment epithelium (RPE) cells. MCMV underwent ocular latency, which was associated with expression of multiple virus genes and from which MCMV could be reactivated by immunosuppression. Latent ocular infection was associated with significant up-regulation of several inflammatory/angiogenic factors. Retinal and choroidal pathologies developed in a progressive manner, with deposits appearing at both basal and apical aspects of the RPE, RPE/choroidal atrophy, photoreceptor degeneration, and neovascularization. The pathologies induced by long-term ocular MCMV latency share features of previously described human ocular diseases, such as age-related macular degeneration.
Collapse
Affiliation(s)
- Jinxian Xu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia; James and Jean Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Xinglou Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia; James and Jean Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Xinyan Zhang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia; James and Jean Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Brendan Marshall
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia; Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia; James and Jean Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Ophthamology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Diego G Espinosa-Heidmann
- James and Jean Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Ophthamology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Ming Zhang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia; James and Jean Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia.
| |
Collapse
|
4
|
Muhuri M, Maeda Y, Ma H, Ram S, Fitzgerald KA, Tai PW, Gao G. Overcoming innate immune barriers that impede AAV gene therapy vectors. J Clin Invest 2021; 131:143780. [PMID: 33393506 DOI: 10.1172/jci143780] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The field of gene therapy has made considerable progress over the past several years. Adeno-associated virus (AAV) vectors have emerged as promising and attractive tools for in vivo gene therapy. Despite the recent clinical successes achieved with recombinant AAVs (rAAVs) for therapeutics, host immune responses against the vector and transgene product have been observed in numerous preclinical and clinical studies. These outcomes have hampered the advancement of AAV gene therapies, preventing them from becoming fully viable and safe medicines. The human immune system is multidimensional and complex. Both the innate and adaptive arms of the immune system seem to play a concerted role in the response against rAAVs. While most efforts have been focused on the role of adaptive immunity and developing ways to overcome it, the innate immune system has also been found to have a critical function. Innate immunity not only mediates the initial response to the vector, but also primes the adaptive immune system to launch a more deleterious attack against the foreign vector. This Review highlights what is known about innate immune responses against rAAVs and discusses potential strategies to circumvent these pathways.
Collapse
Affiliation(s)
- Manish Muhuri
- Horae Gene Therapy Center.,Department of Microbiology and Physiological Systems.,VIDE Program
| | - Yukiko Maeda
- Horae Gene Therapy Center.,VIDE Program.,Department of Medicine
| | | | - Sanjay Ram
- Division of Infectious Diseases and Immunology
| | | | - Phillip Wl Tai
- Horae Gene Therapy Center.,Department of Microbiology and Physiological Systems.,VIDE Program
| | - Guangping Gao
- Horae Gene Therapy Center.,Department of Microbiology and Physiological Systems.,Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
5
|
Boppana SB, Britt WJ. Recent Approaches and Strategies in the Generation of Anti-human Cytomegalovirus Vaccines. Methods Mol Biol 2021; 2244:403-463. [PMID: 33555597 DOI: 10.1007/978-1-0716-1111-1_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human cytomegalovirus is the largest human herpesvirus and shares many core features of other herpesviruses such as tightly regulated gene expression during genome replication and latency as well as the establishment of lifelong persistence following infection. In contrast to stereotypic clinical syndromes associated with alpha-herpesvirus infections, almost all primary HCMV infections are asymptomatic and acquired early in life in most populations in the world. Although asymptomatic in most individuals, HCMV is a major cause of disease in hosts with deficits in adaptive and innate immunity such as infants who are infected in utero and allograft recipients following transplantation. Congenital HCMV is a commonly acquired infection in the developing fetus that can result in a number of neurodevelopmental abnormalities. Similarly, HCMV is a major cause of disease in allograft recipients in the immediate and late posttransplant period and is thought to be a major contributor to chronic allograft rejection. Even though HCMV induces robust innate and adaptive immune responses, it also encodes a vast array of immune evasion functions that are thought aid in its persistence. Immune correlates of protective immunity that prevent or modify intrauterine HCMV infection remain incompletely defined but are thought to consist primarily of adaptive responses in the pregnant mother, thus making congenital HCMV a potentially vaccine modifiable disease. Similarly, HCMV infection in allograft recipients is often more severe in recipients without preexisting adaptive immunity to HCMV. Thus, there has been a considerable effort to modify HCMV specific immunity in transplant recipient either through active immunization or passive transfer of adaptive effector functions. Although efforts to develop an efficacious vaccine and/or passive immunotherapy to limit HCMV disease have been underway for nearly six decades, most have met with limited success at best. In contrast to previous efforts, current HCMV vaccine development has relied on observations of unique properties of HCMV in hopes of reproducing immune responses that at a minimum will be similar to that following natural infection. However, more recent findings have suggested that immunity following naturally acquired HCMV infection may have limited protective activity and almost certainly, is not sterilizing. Such observations suggest that either the induction of natural immunity must be specifically tailored to generate protective activity or alternatively, that providing targeted passive immunity to susceptible populations could be prove to be more efficacious.
Collapse
Affiliation(s)
- Suresh B Boppana
- Departments of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL, USA.,Departments of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - William J Britt
- Departments of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL, USA. .,Departments of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA. .,Departments of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
6
|
Železnjak J, Lisnić VJ, Popović B, Lisnić B, Babić M, Halenius A, L'Hernault A, Roviš TL, Hengel H, Erhard F, Redwood AJ, Vidal SM, Dölken L, Krmpotić A, Jonjić S. The complex of MCMV proteins and MHC class I evades NK cell control and drives the evolution of virus-specific activating Ly49 receptors. J Exp Med 2019; 216:1809-1827. [PMID: 31142589 PMCID: PMC6683999 DOI: 10.1084/jem.20182213] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/04/2019] [Accepted: 05/07/2019] [Indexed: 11/05/2022] Open
Abstract
Železnjak et al. demonstrate that two MCMV-encoded proteins interact with MHC I molecules, forming an altered-self complex that prevents missing self recognition by increasing specificity for inhibitory Ly49 receptors. This led to the evolution of CMV-specific activating Ly49s. CMVs efficiently target MHC I molecules to avoid recognition by cytotoxic T cells. However, the lack of MHC I on the cell surface renders the infected cell susceptible to NK cell killing upon missing self recognition. To counter this, mouse CMV (MCMV) rescues some MHC I molecules to engage inhibitory Ly49 receptors. Here we identify a new viral protein, MATp1, that is essential for MHC I surface rescue. Rescued altered-self MHC I molecules show increased affinity to inhibitory Ly49 receptors, resulting in inhibition of NK cells despite substantially reduced MHC I surface levels. This enables the virus to evade recognition by licensed NK cells. During evolution, this novel viral immune evasion mechanism could have prompted the development of activating NK cell receptors that are specific for MATp1-modified altered-self MHC I molecules. Our study solves a long-standing conundrum of how MCMV avoids recognition by NK cells, unravels a fundamental new viral immune evasion mechanism, and demonstrates how this forced the evolution of virus-specific activating MHC I–restricted Ly49 receptors.
Collapse
Affiliation(s)
- Jelena Železnjak
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Vanda Juranić Lisnić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Branka Popović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Berislav Lisnić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Marina Babić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,Innate Immunity, German Rheumatism Research Centre, a Leibniz Institute, Berlin, Germany
| | - Anne Halenius
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anne L'Hernault
- Precision Medicine and Genomics, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, UK
| | - Tihana Lenac Roviš
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Hartmut Hengel
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florian Erhard
- Institute of Virology and Immunobiology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Alec J Redwood
- Institute for Respiratory Health, University of Western Australia, Western Australia, Australia
| | - Silvia M Vidal
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada.,McGill Center for Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Lars Dölken
- Institute of Virology and Immunobiology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Astrid Krmpotić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia .,Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
7
|
Shao W, Chen X, Samulski RJ, Hirsch ML, Li C. Inhibition of antigen presentation during AAV gene therapy using virus peptides. Hum Mol Genet 2019; 27:601-613. [PMID: 29272432 DOI: 10.1093/hmg/ddx427] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 12/12/2017] [Indexed: 11/14/2022] Open
Abstract
The clinical trial using adeno-associated virus (AAV) vector delivery of mini-dystrophin in patients with Duchenne Muscular Dystrophy (DMD) demonstrated a cytotoxic lymphocyte (CTL) response targeting the transgene product. These mini-dystrophin-specific T-cells have the potential to clear all transduced muscle, presenting the general gene therapy concern of overcoming the CTL response to foreign proteins that provide therapeutic benefit. In this study, we exploited a natural immunosuppression strategy employed by some viruses that results in CTL evasion only in transduced cells. After transfection of the plasmids encoding viral peptides and ovalbumin, which includes the immune-domain epitope SIINFEKL, several viral small peptides (ICP47 and US6) inhibited the SIINFEKL peptide presentation. A single AAV vector genome that consisted of either transgene AAT fused with SIINFEKL epitope and, separately, ICP47 expressed from different promoters or a single fusion protein with ICP47 linked by a furin cleavage peptide (AATOVA-ICP47) decreased antigen presentation. Compared with AAV/AATOVA in which decreased AAT expression was observed at late time points, persistent transgene expression was obtained after systemic administration of AAV/AATOVA-ICP47 vectors in mice. We extended this strategy to DMD gene therapy. After administration of AAV vector encoding human mini-dystrophin fusion protein with ICP47 into mdx mice, a lower mini-dystrophin-specific CTL response was induced. Importantly, the ICP47 fusion to mini-dystrophin inhibited CTLs mediated cytotoxicity. Although demonstrated herein using AAT and mini-dystrophin transgenes in an AAV context, the collective results have implications for all gene therapy applications resulting in foreign peptides by immune suppression in only genetically modified cells.
Collapse
Affiliation(s)
- Wenwei Shao
- Gene Therapy Center, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiaojing Chen
- Gene Therapy Center, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Richard J Samulski
- Gene Therapy Center, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew L Hirsch
- Gene Therapy Center, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Ophthalmology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chengwen Li
- Gene Therapy Center, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
8
|
Almanan M, Raynor J, Sholl A, Wang M, Chougnet C, Cardin RD, Hildeman DA. Tissue-specific control of latent CMV reactivation by regulatory T cells. PLoS Pathog 2017; 13:e1006507. [PMID: 28796839 PMCID: PMC5552023 DOI: 10.1371/journal.ppat.1006507] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 07/02/2017] [Indexed: 12/30/2022] Open
Abstract
Cytomegalovirus (CMV) causes a persistent, lifelong infection. CMV persists in a latent state and undergoes intermittent subclinical viral reactivation that is quelled by ongoing T cell responses. While T cells are critical to maintain control of infection, the immunological factors that promote CMV persistence remain unclear. Here, we investigated the role of regulatory T cells (Treg) in a mouse model of latent CMV infection using Foxp3-diphtheria toxin receptor (Foxp3-DTR) mice. Eight months after infection, MCMV had established latency in the spleen, salivary gland, lung, and pancreas, which was accompanied by an increased frequency of Treg. Administration of diphtheria toxin (DT) after establishment of latency efficiently depleted Treg and drove a significant increase in the numbers of functional MCMV-specific CD4+ and CD8+ T cells. Strikingly, Treg depletion decreased the number of animals with reactivatable latent MCMV in the spleen. Unexpectedly, in the same animals, ablation of Treg drove a significant increase in viral reactivation in the salivary gland that was accompanied with augmented local IL-10 production by Foxp3-CD4+T cells. Further, neutralization of IL-10 after Treg depletion significantly decreased viral load in the salivary gland. Combined, these data show that Treg have divergent control of MCMV infection depending upon the tissue. In the spleen, Treg antagonize CD8+ effector function and promote viral persistence while in the salivary gland Treg prevent IL-10 production and limit viral reactivation and replication. These data provide new insights into the organ-specific roles of Treg in controlling the reactivation of latent MCMV infection.
Collapse
Affiliation(s)
- Maha Almanan
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Immunobiology, Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Jana Raynor
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Immunobiology, Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Allyson Sholl
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Immunobiology, Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Mei Wang
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Infectious Diseases, Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Claire Chougnet
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Immunobiology, Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Rhonda D. Cardin
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Infectious Diseases, Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States of America
| | - David A. Hildeman
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Immunobiology, Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| |
Collapse
|
9
|
Zhao XY, Luo XY, Yu XX, Zhao XS, Han TT, Chang YJ, Huo MR, Xu LP, Zhang XH, Liu KY, Li D, Jiang ZF, Huang XJ. Recipient-donor KIR ligand matching prevents CMV reactivation post-haploidentical T cell-replete transplantation. Br J Haematol 2017; 177:766-781. [PMID: 28466469 DOI: 10.1111/bjh.14622] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/10/2016] [Indexed: 02/01/2023]
Abstract
Licensed natural killer (NK) cells have been demonstrated to have anti-cytomegalovirus (CMV) activity. We prospectively analysed the human leucocyte antigen typing of donor-recipient pairs and the killer cell immunoglobulin-like receptor (KIR) typing of donors for 180 leukaemia patients to assess the predictive roles of licensed NK cells on CMV reactivation post-T-cell-replete haploidentical stem cell transplantation. Multivariate analysis showed that donor-recipient KIR ligand graft-versus-host or host-versus-graft direction mismatch was associated with increased refractory CMV infection (Hazard ratio = 2·556, 95% confidence interval, 1·377-4·744, P = 0·003) post-transplantation. Donor-recipient KIR ligand matching decreased CMV reactivation [51·65% (46·67, 56·62%) vs. 75·28% (70·87, 79·69%), P = 0·012], refractory CMV infection [17·58% (13·77, 21·40%) vs. 35·96% (31·09, 40·82%), P = 0·004] and CMV disease [3·30% (1·51, 5·08%) vs. 11·24% (8·04, 14·43%), P = 0·024] by day 100 post-transplantation. In addition, the percentage of γ-interferon expression on donor-derived NK cells was significantly higher in the recipients among the recipient-donor pairs with a KIR ligand match compared with that in the recipients among the pairs with a KIR ligand graft-versus-host or host-versus-graft direction mismatch on days 30 and 100 post-transplantation (P = 0·036 and 0·047, respectively). These findings have suggested that donor-recipient KIR ligand matching might promote the NK cell licensing process, thereby increasing NK cell-mediated protection against CMV reactivation.
Collapse
Affiliation(s)
- Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China
| | - Xue-Yi Luo
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China
| | - Xing-Xing Yu
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China.,Peking-Tsinghua Centre for Life Sciences, Beijing, China
| | - Xiao-Su Zhao
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China
| | - Ting-Ting Han
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China
| | - Ying-Jun Chang
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China
| | - Ming-Rui Huo
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China
| | - Dan Li
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China
| | - Zheng-Fan Jiang
- Peking-Tsinghua Centre for Life Sciences, Beijing, China.,State Key Laboratory of Protein and Plant Gene Research, Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China.,Peking-Tsinghua Centre for Life Sciences, Beijing, China
| |
Collapse
|
10
|
Zeleznjak J, Popovic B, Krmpotic A, Jonjic S, Lisnic VJ. Mouse cytomegalovirus encoded immunoevasins and evolution of Ly49 receptors - Sidekicks or enemies? Immunol Lett 2017; 189:40-47. [PMID: 28414184 DOI: 10.1016/j.imlet.2017.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/04/2017] [Accepted: 04/11/2017] [Indexed: 12/26/2022]
Abstract
Cytomegaloviruses (CMVs) have dedicated a large portion of their genome towards immune evasion targeting many aspects of the host immune system, particularly NK cells. However, the host managed to cope with the infection by developing multiple mechanisms to recognize viral threat and counterattack it, thus illustrating never-ending evolutionary interplay between CMV and its host. In this review, we will focus on several mechanisms of NK cell evasion by mouse CMV (MCMV), the role of host inhibitory and activating Ly49 receptors involved in the virus control and acquisition of adaptive features by NK cells as a consequence of MCMV infection.
Collapse
Affiliation(s)
- Jelena Zeleznjak
- Department for Histology and Embryology, Faculty of Medicine, University of Rijeka, Croatia
| | - Branka Popovic
- Department for Histology and Embryology, Faculty of Medicine, University of Rijeka, Croatia
| | - Astrid Krmpotic
- Department for Histology and Embryology, Faculty of Medicine, University of Rijeka, Croatia
| | - Stipan Jonjic
- Department for Histology and Embryology, Faculty of Medicine, University of Rijeka, Croatia; Center for Proteomics, Faculty of Medicine, University of Rijeka, Croatia
| | - Vanda Juranic Lisnic
- Department for Histology and Embryology, Faculty of Medicine, University of Rijeka, Croatia; Center for Proteomics, Faculty of Medicine, University of Rijeka, Croatia.
| |
Collapse
|
11
|
Arapović M, Brizić I, Popović B, Jurković S, Jordan S, Krmpotić A, Arapović J, Jonjić S. Intrinsic Contribution of Perforin to NK-Cell Homeostasis during Mouse Cytomegalovirus Infection. Front Immunol 2016; 7:133. [PMID: 27092144 PMCID: PMC4822405 DOI: 10.3389/fimmu.2016.00133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/24/2016] [Indexed: 12/18/2022] Open
Abstract
In addition to their role as effector cells in virus control, natural killer (NK) cells have an immunoregulatory function in shaping the antiviral T-cell response. This function is further pronounced in perforin-deficient mice that show the enhanced NK-cell proliferation and cytokine secretion upon mouse cytomegalovirus (MCMV) infection. Here, we confirmed that stronger activation and maturation of NK cells in perforin-deficient mice correlates with higher MCMV load. To further characterize the immunoregulatory potential of perforin, we compared the response of NK cells that express or do not express perforin using bone-marrow chimeras. Our results demonstrated that the enhanced proliferation and maturation of NK cells in MCMV-infected bone-marrow chimeras is an intrinsic property of perforin-deficient NK cells. Thus, in addition to confirming that NK-cell proliferation is virus load dependent, our data extend this notion demonstrating that perforin plays an intrinsic role as a feedback mechanism in the regulation of NK-cell proliferation during viral infections.
Collapse
Affiliation(s)
- Maja Arapović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka , Rijeka , Croatia
| | - Ilija Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka , Rijeka , Croatia
| | - Branka Popović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka , Rijeka , Croatia
| | - Slaven Jurković
- Department of Medical Physics, Clinical Hospital Rijeka , Rijeka , Croatia
| | - Stefan Jordan
- Department of Oncological Sciences, Tisch Cancer Institute and the Immunology Institute, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| | - Astrid Krmpotić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka , Rijeka , Croatia
| | - Jurica Arapović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia; Faculty of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina
| | - Stipan Jonjić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia; Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
12
|
Cytomegalovirus immune evasion by perturbation of endosomal trafficking. Cell Mol Immunol 2014; 12:154-69. [PMID: 25263490 PMCID: PMC4654299 DOI: 10.1038/cmi.2014.85] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/15/2014] [Accepted: 08/16/2014] [Indexed: 12/30/2022] Open
Abstract
Cytomegaloviruses (CMVs), members of the herpesvirus family, have evolved a variety of mechanisms to evade the immune response to survive in infected hosts and to establish latent infection. They effectively hide infected cells from the effector mechanisms of adaptive immunity by eliminating cellular proteins (major histocompatibility Class I and Class II molecules) from the cell surface that display viral antigens to CD8 and CD4 T lymphocytes. CMVs also successfully escape recognition and elimination of infected cells by natural killer (NK) cells, effector cells of innate immunity, either by mimicking NK cell inhibitory ligands or by downregulating NK cell-activating ligands. To accomplish these immunoevasion functions, CMVs encode several proteins that function in the biosynthetic pathway by inhibiting the assembly and trafficking of cellular proteins that participate in immune recognition and thereby, block their appearance at the cell surface. However, elimination of these proteins from the cell surface can also be achieved by perturbation of their endosomal route and subsequent relocation from the cell surface into intracellular compartments. Namely, the physiological route of every cellular protein, including immune recognition molecules, is characterized by specific features that determine its residence time at the cell surface. In this review, we summarize the current understanding of endocytic trafficking of immune recognition molecules and perturbations of the endosomal system during infection with CMVs and other members of the herpesvirus family that contribute to their immune evasion mechanisms.
Collapse
|
13
|
Brizić I, Lenac Roviš T, Krmpotić A, Jonjić S. MCMV avoidance of recognition and control by NK cells. Semin Immunopathol 2014; 36:641-50. [DOI: 10.1007/s00281-014-0441-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 07/15/2014] [Indexed: 01/27/2023]
|
14
|
Jost NH, Abel S, Hutzler M, Sparwasser T, Zimmermann A, Roers A, Müller W, Klopfleisch R, Hengel H, Westendorf AM, Buer J, Hansen W. Regulatory T cells and T‐cell‐derived IL‐10 interfere with effective anti‐cytomegalovirus immune response. Immunol Cell Biol 2014; 92:860-71. [DOI: 10.1038/icb.2014.62] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/25/2014] [Accepted: 06/26/2014] [Indexed: 02/07/2023]
Affiliation(s)
- Nils H Jost
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg‐EssenEssenGermany
| | - Simone Abel
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg‐EssenEssenGermany
| | - Marina Hutzler
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg‐EssenEssenGermany
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCOREHannoverGermany
| | | | - Axel Roers
- Institute for Immunology, TU DresdenDresdenGermany
| | - Werner Müller
- Faculty of Life Sciences, University of ManchesterManchesterUK
| | | | - Hartmut Hengel
- Institute for Virology, University Medical Center Freiburg, Albert‐Ludwigs UniversityFreiburgGermany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg‐EssenEssenGermany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg‐EssenEssenGermany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg‐EssenEssenGermany
- Institute for Virology, University Medical Center Freiburg, Albert‐Ludwigs UniversityFreiburgGermany
| |
Collapse
|
15
|
Alexandre YO, Cocita CD, Ghilas S, Dalod M. Deciphering the role of DC subsets in MCMV infection to better understand immune protection against viral infections. Front Microbiol 2014; 5:378. [PMID: 25120535 PMCID: PMC4114203 DOI: 10.3389/fmicb.2014.00378] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/04/2014] [Indexed: 12/22/2022] Open
Abstract
Infection of mice with murine cytomegalovirus (MCMV) recapitulates many physiopathological characteristics of human CMV infection and enables studying the interactions between a virus and its natural host. Dendritic cells (DC) are mononuclear phagocytes linking innate and adaptive immunity which are both necessary for MCMV control. DC are critical for the induction of cellular immunity because they are uniquely efficient for the activation of naïve T cells during their first encounter with a pathogen. DC are equipped with a variety of innate immune recognition receptors (I2R2) allowing them to detect pathogens or infections and to engulf molecules, microorganisms or cellular debris. The combinatorial engagement of I2R2 during infections controls DC maturation and shapes their response in terms of cytokine production, activation of natural killer (NK) cells and functional polarization of T cells. Several DC subsets exist which express different arrays of I2R2 and are specialized in distinct functions. The study of MCMV infection helped deciphering the physiological roles of DC subsets and their molecular regulation. It allowed the identification and first in vivo studies of mouse plasmacytoid DC which produce high level of interferons-α/β early after infection. Despite its ability to infect DC and dampen their functions, MCMV induces very robust, efficient and long-lasting CD8 T cell responses. Their priming may rely on the unique ability of uninfected XCR1+ DC to cross-present engulfed viral antigens and thus to counter MCMV interference with antigen presentation. A balance appears to have been reached during co-evolution, allowing controlled replication of the virus for horizontal spread without pathological consequences for the immunocompetent host. We will discuss the role of the interplay between the virus and DC in setting this balance, and how advancing this knowledge further could help develop better vaccines against other intracellular infectious agents.
Collapse
Affiliation(s)
- Yannick O Alexandre
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| | - Clément D Cocita
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| | - Sonia Ghilas
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| | - Marc Dalod
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| |
Collapse
|
16
|
Forbes CA, Scalzo AA, Degli-Esposti MA, Coudert JD. Ly49C-dependent control of MCMV Infection by NK cells is cis-regulated by MHC Class I molecules. PLoS Pathog 2014; 10:e1004161. [PMID: 24873973 PMCID: PMC4038614 DOI: 10.1371/journal.ppat.1004161] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 04/20/2014] [Indexed: 11/18/2022] Open
Abstract
Natural Killer (NK) cells are crucial in early resistance to murine cytomegalovirus (MCMV) infection. In B6 mice, the activating Ly49H receptor recognizes the viral m157 glycoprotein on infected cells. We previously identified a mutant strain (MCMVG1F) whose variant m157 also binds the inhibitory Ly49C receptor. Here we show that simultaneous binding of m157 to the two receptors hampers Ly49H-dependent NK cell activation as Ly49C-mediated inhibition destabilizes NK cell conjugation with their targets and prevents the cytoskeleton reorganization that precedes killing. In B6 mice, as most Ly49H+ NK cells do not co-express Ly49C, the overall NK cell response remains able to control MCMVm157G1F infection. However, in B6 Ly49C transgenic mice where all NK cells express the inhibitory receptor, MCMV infection results in altered NK cell activation associated with increased viral replication. Ly49C-mediated inhibition also regulates Ly49H-independent NK cell activation. Most interestingly, MHC class I regulates Ly49C function through cis-interactions that mask the receptor and restricts m157 binding. B6 Ly49C Tg, β2m ko mice, whose Ly49C receptors are unmasked due to MHC class I deficient expression, are highly susceptible to MCMVm157G1F and are unable to control a low-dose infection. Our study provides novel insights into the mechanisms that regulate NK cell activation during viral infection.
Collapse
Affiliation(s)
- Catherine A. Forbes
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Anthony A. Scalzo
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Mariapia A. Degli-Esposti
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
- Centre for Ophthalmology and Vision Science, M517, University of Western Australia, Crawley, Western Australia, Australia
| | - Jerome D. Coudert
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
- Centre for Ophthalmology and Vision Science, M517, University of Western Australia, Crawley, Western Australia, Australia
- * E-mail:
| |
Collapse
|
17
|
Reuben A, Phénix M, Santos MM, Lapointe R. The WT hemochromatosis protein HFE inhibits CD8⁺ T-lymphocyte activation. Eur J Immunol 2014; 44:1604-14. [PMID: 24643698 DOI: 10.1002/eji.201343955] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 01/21/2014] [Accepted: 02/11/2014] [Indexed: 11/05/2022]
Abstract
MHC class I (MHC I) antigen presentation is a ubiquitous process by which cells present endogenous proteins to CD8(+) T lymphocytes during immune surveillance and response. Hereditary hemochromatosis protein, HFE, is involved in cellular iron uptake but, while structurally homologous to MHC I, is unable to bind peptides. However, increasing evidence suggests a role for HFE in the immune system. Here, we investigated the impact of HFE on CD8(+) T-lymphocyte activation. Using transient HFE transfection assays in a model of APCs, we show that WT HFE (HFEWT ), but not C282Y-mutated HFE, inhibits secretion of MIP-1β from antigen-specific CD8(+) T lymphocytes. HFEWT expression also resulted in major decreases in CD8(+) T-lymphocyte activation as measured by 4-1BB expression. We further demonstrate that inhibition of CD8(+) T-lymphocyte activation was independent of MHC I surface levels, β2-m competition, HFE interaction with transferrin receptor, antigen origin, or epitope affinity. Finally, we identified the α1-2 domains of HFEWT as being responsible for inhibiting CD8(+) T-lymphocyte activation. Our data imply a new role for HFEWT in altering CD8(+) T-lymphocyte reactivity, which could modulate antigen immunogenicity.
Collapse
Affiliation(s)
- Alexandre Reuben
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) Institut du cancer de Montréal, Montréal, Québec, Canada; Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | | | | | | |
Collapse
|
18
|
Recent approaches and strategies in the generation of antihuman cytomegalovirus vaccines. Methods Mol Biol 2014; 1119:311-48. [PMID: 24639230 DOI: 10.1007/978-1-62703-788-4_17] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The development of prophylactic and to lesser extent therapeutic vaccines for the prevention of disease associated with human cytomegalovirus (HCMV) infections has received considerable attention from biomedical researchers and pharmaceutical companies over the previous 15 years, even though attempts to produce such vaccines have been described in the literature for over 40 years. Studies of the natural history of congenital HCMV infection and infection in allograft recipients have suggested that prophylaxis of disease associated with HCMV infection could be possible, particularly in hosts at risk for more severe disease secondary to the lack of preexisting immunity. Provided a substantial understanding of immune response to HCMV together with several animal models that faithfully recapitulate aspects of human infection and immunity, investigators seem well positioned to design and test candidate vaccines. Yet more recent studies of the role of a maternal immunity in the natural history of congenital HCMV infection, including the recognition that reinfection of previously immune women by genetically distinct strains of HCMV occur in populations with a high seroprevalence, have raised several questions about the nature of protective immunity in maternal populations. This finding coupled with observations that have documented a significant incidence of damaging congenital infections in offspring of women with immunity to HCMV prior to conception has suggested that vaccine development based on conventional paradigms of adaptive immunity to viral infections may be of limited value in the prevention of damaging congenital HCMV infections. Perhaps a more achievable goal will be prophylactic vaccines to modify HCMV associated disease in allograft transplant recipients. Although recent descriptions of the results from vaccine trials have been heralded as evidence of an emerging success in the quest for a HCMV vaccine, careful analyses of these studies have also revealed that major hurdles remain to be addressed by current strategies.
Collapse
|
19
|
Miletić A, Krmpotić A, Jonjić S. The evolutionary arms race between NK cells and viruses: who gets the short end of the stick? Eur J Immunol 2013; 43:867-77. [PMID: 23440773 DOI: 10.1002/eji.201243101] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 02/11/2013] [Accepted: 02/21/2013] [Indexed: 01/17/2023]
Abstract
NK cells are innate lymphocytes that play a key role in the control of various viral infections. Recent studies indicate that NK cells may acquire some features of adaptive immune cells, including the formation of long-lived memory cells. A large and growing body of data indicates that NK cells regulate the adaptive immune response as well. The function and the activation status of NK cells are tightly regulated by signals induced by a broad range of inhibitory and activating cell surface receptors and cytokines released by other immune cells. Here, we review the function of mouse NK-cell receptors involved in virus control and in the regulation of the adaptive immune response. In addition, we discuss viral strategies used to evade NK-cell-mediated control during infection. Finally, the role of several activating Ly49 receptors specific for mouse cytomegalovirus (MCMV), as well as some controversial issues in the field, will be discussed.
Collapse
Affiliation(s)
- Antonija Miletić
- Department of Histology and Embryology, Faculty of Medicine, Rijeka, Croatia
| | | | | |
Collapse
|
20
|
Fleming P, Kvansakul M, Voigt V, Kile BT, Kluck RM, Huang DCS, Degli-Esposti MA, Andoniou CE. MCMV-mediated inhibition of the pro-apoptotic Bak protein is required for optimal in vivo replication. PLoS Pathog 2013; 9:e1003192. [PMID: 23468630 PMCID: PMC3585157 DOI: 10.1371/journal.ppat.1003192] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 12/28/2012] [Indexed: 01/29/2023] Open
Abstract
Successful replication and transmission of large DNA viruses such as the cytomegaloviruses (CMV) family of viruses depends on the ability to interfere with multiple aspects of the host immune response. Apoptosis functions as a host innate defence mechanism against viral infection, and the capacity to interfere with this process is essential for the replication of many viruses. The Bcl-2 family of proteins are the principle regulators of apoptosis, with two pro-apoptotic members, Bax and Bak, essential for apoptosis to proceed. The m38.5 protein encoded by murine CMV (MCMV) has been identified as Bax-specific inhibitor of apoptosis. Recently, m41.1, a protein product encoded by the m41 open reading frame (ORF) of MCMV, has been shown to inhibit Bak activity in vitro. Here we show that m41.1 is critical for optimal MCMV replication in vivo. Growth of a m41.1 mutant was attenuated in multiple organs, a defect that was not apparent in Bak−/− mice. Thus, m41.1 promotes MCMV replication by inhibiting Bak-dependent apoptosis during in vivo infection. The results show that Bax and Bak mediate non-redundant functions during MCMV infection and that the virus produces distinct inhibitors for each protein to counter the activity of these proteins. The cytomegaloviruses (CMV) are a family of viruses that establish a latent infection that lasts for the life of the host, with the virus able to reactivate when the host is immunosuppressed. We have used murine CMV (MCMV) as a model to understand how CMV interferes with the anti-viral immune response. Apoptosis, or programmed cell death, is one of the defence mechanisms used by multicellular organisms to impair viral infection. In order for viral replication to proceed, many viruses have evolved mechanisms to prevent the apoptosis of infected host cells. Under most circumstances the activation of Bax, or the closely related protein Bak, is required for apoptosis to proceed. The m41.1 protein was recently identified as a candidate Bak inhibitor during in vitro infection. We have generated a mutant virus which is unable to produce the m41.1 protein and found that growth of this virus was attenuated in wild-type mice. Importantly, growth of the mutant virus was equivalent to that of the wild-type virus in mice lacking the Bak protein. These studies establish that m41.1 is an inhibitor of Bak and that the capacity to prevent apoptosis triggered by Bak is required for efficient replication of MCMV in vivo.
Collapse
Affiliation(s)
- Peter Fleming
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Marc Kvansakul
- Department of Biochemistry, La Trobe University, Melbourne, Victoria, Australia
| | - Valentina Voigt
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Benjamin T. Kile
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Ruth M. Kluck
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - David C. S. Huang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Mariapia A. Degli-Esposti
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Christopher E. Andoniou
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
- * E-mail:
| |
Collapse
|
21
|
Cytomegalovirus impairs antiviral CD8+ T cell immunity by recruiting inflammatory monocytes. Immunity 2012; 37:122-33. [PMID: 22840843 DOI: 10.1016/j.immuni.2012.04.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 04/06/2012] [Accepted: 04/19/2012] [Indexed: 12/24/2022]
Abstract
Inflammatory monocytes are key early responders to infection that contribute to pathogen-host interactions in diverse ways. Here, we report that the murine cytomegalovirus-encoded CC chemokine, MCK2, enhanced CCR2-dependent recruitment of these cells to modulate antiviral immunity, impairing virus-specific CD8(+) T cell expansion and differentiation into effector cytotoxic T lymphocytes, thus reducing the capacity to eliminate viral antigen-bearing cells and slowing viral clearance. Adoptive transfer of inflammatory monocytes into Ccr2(-/-)Ccl2(-/-) mice impaired virus antigen-specific clearance. Cytomegalovirus therefore enhances a natural CCR2-dependent immune regulatory network to modulate adaptive immunity via nitric oxide production, reminiscent of the monocytic subtype of myeloid-derived suppressor cells primarily implicated in cancer immunomodulation.
Collapse
|
22
|
Murine cytomegalovirus immune evasion proteins operative in the MHC class I pathway of antigen processing and presentation: state of knowledge, revisions, and questions. Med Microbiol Immunol 2012; 201:497-512. [PMID: 22961127 DOI: 10.1007/s00430-012-0257-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 08/22/2012] [Indexed: 12/15/2022]
Abstract
Medical interest in cytomegalovirus (CMV) is based on lifelong neurological sequelae, such as sensorineural hearing loss and mental retardation, resulting from congenital infection of the fetus in utero, as well as on CMV disease with multiple organ manifestations and graft loss in recipients of hematopoietic cell transplantation or solid organ transplantation. CMV infection of transplantation recipients occurs consequent to reactivation of virus harbored in a latent state in the transplanted donor cells and tissues, or in the tissues of the transplantation recipient herself or himself. Hence, CMV infection is a paradigm for a viral infection that causes disease primarily in the immunocompromised host, while infection of the immunocompetent host is associated with only mild and nonspecific symptoms so that it usually goes unnoticed. Thus, CMV is kept under strict immune surveillance. These medical facts are in apparent conflict with the notion that CMVs in general, human CMV as well as animal CMVs, are masters of 'immune evasion', which during virus-host co-speciation have convergently evolved sophisticated mechanisms to avoid their recognition by innate and adaptive immunity of their respective host species, with viral genes apparently dedicated to serve just this purpose (Reddehase in Nat Rev Immunol 2:831-844, 2002). With focus on viral interference with antigen presentation to CD8 T cells in the preclinical model of murine CMV infection, we try here to shed some more light on the in vivo balance between host immune surveillance of CMV infection and viral 'immune evasion' strategies.
Collapse
|
23
|
All is fair in virus-host interactions: NK cells and cytomegalovirus. Trends Mol Med 2011; 17:677-85. [PMID: 21852192 DOI: 10.1016/j.molmed.2011.07.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 07/11/2011] [Accepted: 07/12/2011] [Indexed: 12/12/2022]
Abstract
The infection of mice with mouse cytomegalovirus (MCMV) as a model of human cytomegalovirus (HCMV) infection has been particularly informative in elucidating the role of innate and adaptive immune response mechanisms during infection. Millions of years of co-evolution between cytomegaloviruses (CMV) and their hosts has resulted in numerous attempts to overwhelm each other. CMVs devote many genes to modulating the host natural killer (NK) cell response and NK cells employ many strategies to cope with CMV infection. While focusing on these attack-counterattack measures, this review will discuss several novel mechanisms of immune evasion by MCMV, the role of Ly49 receptors in mediating resistance to MCMV, and the impact of the initial NK cell response on the shaping of adaptive immunity.
Collapse
|
24
|
How the virus outsmarts the host: function and structure of cytomegalovirus MHC-I-like molecules in the evasion of natural killer cell surveillance. J Biomed Biotechnol 2011; 2011:724607. [PMID: 21765638 PMCID: PMC3134397 DOI: 10.1155/2011/724607] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 03/28/2011] [Indexed: 12/18/2022] Open
Abstract
Natural killer (NK) cells provide an initial host immune response to infection by many viral pathogens. Consequently, the viruses have evolved mechanisms to attenuate the host response, leading to improved viral fitness. One mechanism employed by members of the β-herpesvirus family, which includes the cytomegaloviruses, is to modulate the expression of cell surface ligands recognized by NK cell activation molecules. A novel set of cytomegalovirus (CMV) genes, exemplified by the mouse m145 family, encode molecules that have structural and functional features similar to those of host major histocompatibility-encoded (MHC) class I molecules, some of which are known to contribute to immune evasion. In this review, we explore the function, structure, and evolution of MHC-I-like molecules of the CMVs and speculate on the dynamic development of novel immunoevasive functions based on the MHC-I protein fold.
Collapse
|
25
|
Lacaze P, Forster T, Ross A, Kerr LE, Salvo-Chirnside E, Lisnic VJ, López-Campos GH, García-Ramírez JJ, Messerle M, Trgovcich J, Angulo A, Ghazal P. Temporal profiling of the coding and noncoding murine cytomegalovirus transcriptomes. J Virol 2011; 85:6065-76. [PMID: 21471238 PMCID: PMC3126304 DOI: 10.1128/jvi.02341-10] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 03/28/2011] [Indexed: 12/20/2022] Open
Abstract
The global transcriptional program of murine cytomegalovirus (MCMV), involving coding, noncoding, and antisense transcription, remains unknown. Here we report an oligonucleotide custom microarray platform capable of measuring both coding and noncoding transcription on a genome-wide scale. By profiling MCMV wild-type and immediate-early mutant strains in fibroblasts, we found rapid activation of the transcriptome by 6.5 h postinfection, with absolute dependency on ie3, but not ie1 or ie2, for genomic programming of viral gene expression. Evidence is also presented to show, for the first time, genome-wide noncoding and bidirectional transcription at late stages of MCMV infection.
Collapse
Affiliation(s)
- Paul Lacaze
- Division of Pathway Medicine, The University of Edinburgh, The Chancellor's Building, College of Medicine, 49 Little France Crescent, Edinburgh, United Kingdom
| | - Thorsten Forster
- Division of Pathway Medicine, The University of Edinburgh, The Chancellor's Building, College of Medicine, 49 Little France Crescent, Edinburgh, United Kingdom
| | - Alan Ross
- Division of Pathway Medicine, The University of Edinburgh, The Chancellor's Building, College of Medicine, 49 Little France Crescent, Edinburgh, United Kingdom
| | - Lorraine E. Kerr
- Centre for Systems Biology at Edinburgh, The University of Edinburgh, Darwin Building, King's Buildings Campus, Mayfield Road, Edinburgh, United Kingdom
| | - Eliane Salvo-Chirnside
- Centre for Systems Biology at Edinburgh, The University of Edinburgh, Darwin Building, King's Buildings Campus, Mayfield Road, Edinburgh, United Kingdom
| | - Vanda Juranic Lisnic
- Department of Histology and Embryology, Faculty of Medicine, Rijeka University, Croatia
| | | | - José J. García-Ramírez
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla—La Mancha, Avenida de Almansa 14, 02006 Albacete, Spain
| | - Martin Messerle
- Department of Virology, Hannover Medical School, Hannover, Germany
| | - Joanne Trgovcich
- Department of Pathology, The Ohio State University, Columbus, Ohio 43210
| | - Ana Angulo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Peter Ghazal
- Division of Pathway Medicine, The University of Edinburgh, The Chancellor's Building, College of Medicine, 49 Little France Crescent, Edinburgh, United Kingdom
- Centre for Systems Biology at Edinburgh, The University of Edinburgh, Darwin Building, King's Buildings Campus, Mayfield Road, Edinburgh, United Kingdom
| |
Collapse
|
26
|
The role of cell types in cytomegalovirus infection in vivo. Eur J Cell Biol 2011; 91:70-7. [PMID: 21492952 DOI: 10.1016/j.ejcb.2011.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Revised: 02/09/2011] [Accepted: 02/14/2011] [Indexed: 11/20/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the major viral cause of morbidity in immune compromised patients and of pre- and perinatal pathology in newborns. The clinical manifestations are highly variable and the principles which govern these differences cannot be understood without detailed knowledge on tissue specific aspects of HCMV infection. For decades the role of individual cell types during cytomegalovirus infection and disease has been discussed. The pathogenesis of mouse cytomegalovirus (MCMV) mirrors the human infection in many aspects. Although only MCMV infection is studied extensively at the level of organs, the relative contribution of specific cell types to viral pathogenesis in vivo has remained enigmatic. Here we discuss new approaches based on the cre/loxP-system to label nascent virus progeny or to lift a replication block. The salient aspect of this technique is the change of viral genome properties selectively in cells that express cre during infection in vivo. This allowed us to study the role of endothelial cells and hepatocytes for virus dissemination and will permit detailed studies on innate and adaptive immune responses to CMV.
Collapse
|
27
|
Babić M, Pyzik M, Zafirova B, Mitrović M, Butorac V, Lanier LL, Krmpotić A, Vidal SM, Jonjić S. Cytomegalovirus immunoevasin reveals the physiological role of "missing self" recognition in natural killer cell dependent virus control in vivo. J Exp Med 2010; 207:2663-73. [PMID: 21078887 PMCID: PMC2989764 DOI: 10.1084/jem.20100921] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 10/18/2010] [Indexed: 12/14/2022] Open
Abstract
Cytomegaloviruses (CMVs) are renowned for interfering with the immune system of their hosts. To sidestep antigen presentation and destruction by CD8(+) T cells, these viruses reduce expression of major histocompatibility complex class I (MHC I) molecules. However, this process sensitizes the virus-infected cells to natural killer (NK) cell-mediated killing via the "missing self" axis. Mouse cytomegalovirus (MCMV) uses m152 and m06 encoded proteins to inhibit surface expression of MHC I molecules. In addition, it encodes another protein, m04, which forms complexes with MHC I and escorts them to the cell surface. This mechanism is believed to prevent NK cell activation and killing by restoring the "self" signature and allowing the engagement of inhibitory Ly49 receptors on NK cells. Here we show that MCMV lacking m04 was attenuated in an NK cell- and MHC I-dependent manner. NK cell-mediated control of the infection was dependent on the presence of NK cell subsets expressing different inhibitory Ly49 receptors. In addition to providing evidence for immunoevasion strategies used by CMVs to avoid NK cell control via the missing-self pathway, our study is the first to demonstrate that missing self-dependent NK cell activation is biologically relevant in the protection against viral infection in vivo.
Collapse
Affiliation(s)
- Marina Babić
- Department of Histology and Embryology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Michal Pyzik
- Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada
| | - Biljana Zafirova
- Department of Histology and Embryology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Maja Mitrović
- Department of Histology and Embryology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Višnja Butorac
- Department of Histology and Embryology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Lewis L. Lanier
- Department of Microbiology and Immunology and the Cancer Research Institute, University of California, San Francisco, San Francisco, CA 94115
| | - Astrid Krmpotić
- Department of Histology and Embryology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Silvia M. Vidal
- Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada
| | - Stipan Jonjić
- Department of Histology and Embryology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
28
|
Murine cytomegalovirus perturbs endosomal trafficking of major histocompatibility complex class I molecules in the early phase of infection. J Virol 2010; 84:11101-12. [PMID: 20719942 DOI: 10.1128/jvi.00988-10] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Murine cytomegalovirus (MCMV) functions interfere with protein trafficking in the secretory pathway. In this report we used Δm138-MCMV, a recombinant virus with a deleted viral Fc receptor, to demonstrate that MCMV also perturbs endosomal trafficking in the early phase of infection. This perturbation had a striking impact on cell surface-resident major histocompatibility complex class I (MHC-I) molecules due to the complementary effect of MCMV immunoevasins, which block their egress from the secretory pathway. In infected cells, constitutively endocytosed cell surface-resident MHC-I molecules were arrested and retained in early endosomal antigen 1 (EEA1)-positive and lysobisphosphatidic acid (LBPA)-negative perinuclear endosomes together with clathrin-dependent cargo (transferrin receptor, Lamp1, and epidermal growth factor receptor). Their progression from these endosomes into recycling and degradative routes was inhibited. This arrest was associated with a reduction of the intracellular content of Rab7 and Rab11, small GTPases that are essential for the maturation of recycling and endolysosomal domains of early endosomes. The reduced recycling of MHC-I in Δm138-MCMV-infected cells was accompanied by their accelerated loss from the cell surface. The MCMV function that affects cell surface-resident MHC-I was activated in later stages of the early phase of viral replication, after the expression of known immunoevasins. MCMV without the three immunoevasins (the m04, m06, and m152 proteins) encoded a function that affects endosomal trafficking. This function, however, was not sufficient to reduce the cell surface expression of MHC-I in the absence of the transport block in the secretory pathway.
Collapse
|
29
|
Lisnić VJ, Krmpotić A, Jonjić S. Modulation of natural killer cell activity by viruses. Curr Opin Microbiol 2010; 13:530-9. [PMID: 20558100 DOI: 10.1016/j.mib.2010.05.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 05/26/2010] [Accepted: 05/26/2010] [Indexed: 11/27/2022]
Abstract
Since their discovery, our understanding of NK cells has evolved from branding them marginal innate immunity cells to key players in anti-viral and anti-tumor immunity. Importance of NK cells in control of various viral infections is perhaps best illustrated by the existence of plethora of viral mechanisms aimed to modulate their function. These mechanisms include not only virally encoded immunoevasion proteins but also viral miRNA. Moreover, the evidence has been accumulated supporting the role of viral immunoevasion of NK cells in viral pathogenesis in vivo.
Collapse
Affiliation(s)
- Vanda Juranić Lisnić
- Department for Histology and Embryology and Center for Proteomics, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatia
| | | | | |
Collapse
|
30
|
Cunningham PT, Lloyd ML, Harvey NL, Williams E, Hardy CM, Redwood AJ, Lawson MA, Shellam GR. Promoter control over foreign antigen expression in a murine cytomegalovirus vaccine vector. Vaccine 2010; 29:141-51. [PMID: 20338212 DOI: 10.1016/j.vaccine.2010.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 03/03/2010] [Accepted: 03/09/2010] [Indexed: 11/30/2022]
Abstract
Previous studies have reported on the development of a recombinant murine cytomegalovirus (rMCMV) containing the mouse zona pellucida 3 (mZP3) gene for use as a virally vectored immunocontraceptive (VVIC). This study aimed to alter promoter control over foreign antigen expression and cellular localisation of the antigen expressed in order to overcome virus attenuation previously encountered. Early studies reported on the mZP3 gene expressed by a strong constitutive human cytomegalovirus immediate-early 1 promoter (pHCMV IE1). This virus was able to induce >90% infertility in BALB/c mice despite being heavily attenuated in vivo. In this study the mZP3 was placed under the control of the MCMV early 1 (pMCMV E1) promoter and the inducible tetracycline promoter (Tet-On). In both instances the recombinant virus was able to induce infertility in directly infected mice. However, the viruses remained attenuated. This study demonstrated the capacity to manipulate the nature of the immune response by altering promoter control over foreign antigen expression and cellular localisation of the expressed antigen. We were able to demonstrate that by using the MCMV E1 promoter it was still possible to sterilize female BALB/c mice with an MCMV vector expressing mZP3. The use of the MCMV E1 promoter provides an added level of safety to any MCMV based VVIC approach as it only allows for transgene expression in MCMV permissive cells.
Collapse
Affiliation(s)
- Paula T Cunningham
- Discipline of Microbiology and Immunology, School of Biomedical, Biomolecular and Chemical Sciences, University of Western Australia, Crawley, Western Australia 6009, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Orr MT, Lanier LL. Inhibitory Ly49 receptors on mouse natural killer cells. Curr Top Microbiol Immunol 2010; 350:67-87. [PMID: 20680808 DOI: 10.1007/82_2010_85] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Ly49 receptors, which are expressed in a stochastic manner on subsets of murine natural killer (NK) cells, T cells, and other cells, are encoded by the Klra gene family and include receptors with either inhibitory or activating function. All of the inhibitory Ly49 receptors are characterized by an immunoreceptor tyrosine-based inhibitory motif in their cytoplasmic domain, which upon phosphorylation recruits tyrosine or lipid phosphatases to dampen signals transmitted through other activating receptors. Most of the inhibitory Ly49 receptors recognize polymorphic epitopes on major histocompatibility complex (MHC) class I proteins as ligands. Here, we review the polymorphism, ligand specificity, and signaling capacity of the inhibitory Ly49 receptors and discuss how these molecules regulate NK cell development and function.
Collapse
Affiliation(s)
- Mark T Orr
- Department of Microbiology and Immunology and the Cancer Research Institute, University of California, San Francisco, CA, 94143, USA.
| | | |
Collapse
|
32
|
Immune evasion proteins of murine cytomegalovirus preferentially affect cell surface display of recently generated peptide presentation complexes. J Virol 2009; 84:1221-36. [PMID: 19906905 DOI: 10.1128/jvi.02087-09] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
For recognition of infected cells by CD8 T cells, antigenic peptides are presented at the cell surface, bound to major histocompatibility complex class I (MHC-I) molecules. Downmodulation of cell surface MHC-I molecules is regarded as a hallmark function of cytomegalovirus-encoded immunoevasins. The molecular mechanisms by which immunoevasins interfere with the MHC-I pathway suggest, however, that this downmodulation may be secondary to an interruption of turnover replenishment and that hindrance of the vesicular transport of recently generated peptide-MHC (pMHC) complexes to the cell surface is the actual function of immunoevasins. Here we have used the model of murine cytomegalovirus (mCMV) infection to provide experimental evidence for this hypothesis. To quantitate pMHC complexes at the cell surface after infection in the presence and absence of immunoevasins, we generated the recombinant viruses mCMV-SIINFEKL and mCMV-Deltam06m152-SIINFEKL, respectively, expressing the K(b)-presented peptide SIINFEKL with early-phase kinetics in place of an immunodominant peptide of the viral carrier protein gp36.5/m164. The data revealed approximately 10,000 K(b) molecules presenting SIINFEKL in the absence of immunoevasins, which is an occupancy of approximately 10% of all cell surface K(b) molecules, whereas immunoevasins reduced this number to almost the detection limit. To selectively evaluate their effect on preexisting pMHC complexes, cells were exogenously loaded with SIINFEKL peptide shortly after infection with mCMV-SIINFEKA, in which endogenous presentation is prevented by an L174A mutation of the C-terminal MHC-I anchor residue. The data suggest that pMHC complexes present at the cell surface in advance of immunoevasin gene expression are downmodulated due to constitutive turnover in the absence of resupply.
Collapse
|
33
|
Xie X, Stadnisky MD, Brown MG. MHC class I Dk locus and Ly49G2+ NK cells confer H-2k resistance to murine cytomegalovirus. THE JOURNAL OF IMMUNOLOGY 2009; 182:7163-71. [PMID: 19454713 DOI: 10.4049/jimmunol.0803933] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Essential NK cell-mediated murine CMV (MCMV) resistance is under histocompatibility-2(k) (H-2(k)) control in MA/My mice. We generated a panel of intra-H2(k) recombinant strains from congenic C57L.M-H2(k/b) (MCMV resistant) mice for precise genetic mapping of the critical interval. Recombination breakpoint sites were precisely mapped and MCMV resistance/susceptibility traits were determined for each of the new lines to identify the MHC locus. Strains C57L.M-H2(k)(R7) (MCMV resistant) and C57L.M-H2(k)(R2) (MCMV susceptible) are especially informative; we found that allelic variation in a 0.3-megabase interval in the class I D locus confers substantial difference in MCMV control phenotypes. When NK cell subsets responding to MCMV were examined, we found that Ly49G2(+) NK cells rapidly expand and selectively acquire an enhanced capacity for cytolytic functions only in C57L.M-H2(k)(R7). We further show that depletion of Ly49G2(+) NK cells before infection abrogated MCMV resistance in C57L.M-H2(k)(R7). We conclude that the MHC class I D locus prompts expansion and activation of Ly49G2(+) NK cells that are needed in H-2(k) MCMV resistance.
Collapse
Affiliation(s)
- Xuefang Xie
- Department of Microbiology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
34
|
Abstract
The NKG2D receptor is one of the most potent activating natural killer cell receptors involved in antiviral responses. The mouse NKG2D ligands MULT-1, RAE-1, and H60 are regulated by murine cytomegalovirus (MCMV) proteins m145, m152, and m155, respectively. In addition, the m138 protein interferes with the expression of both MULT-1 and H60. We show here that one of five RAE-1 isoforms, RAE-1delta, is resistant to downregulation by MCMV and that this escape has functional importance in vivo. Although m152 retained newly synthesized RAE-1delta and RAE-1gamma in the endoplasmic reticulum, no viral regulator was able to affect the mature RAE-1delta form which remains expressed on the surfaces of infected cells. This differential susceptibility to downregulation by MCMV is not a consequence of faster maturation of RAE-1delta compared to RAE-1gamma but rather an intrinsic property of the mature surface-resident protein. This difference can be attributed to the absence of a PLWY motif from RAE-1delta. Altogether, these findings provide evidence for a novel mechanism of host escape from viral immunoevasion of NKG2D-dependent control.
Collapse
|
35
|
Kielczewska A, Pyzik M, Sun T, Krmpotic A, Lodoen MB, Munks MW, Babic M, Hill AB, Koszinowski UH, Jonjic S, Lanier LL, Vidal SM. Ly49P recognition of cytomegalovirus-infected cells expressing H2-Dk and CMV-encoded m04 correlates with the NK cell antiviral response. ACTA ACUST UNITED AC 2009; 206:515-23. [PMID: 19255146 PMCID: PMC2699136 DOI: 10.1084/jem.20080954] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Natural killer (NK) cells are crucial in resistance to certain viral infections, but the mechanisms used to recognize infected cells remain largely unknown. Here, we show that the activating Ly49P receptor recognizes cells infected with mouse cytomegalovirus (MCMV) by a process that requires the presence of H2-Dk and the MCMV m04 protein. Using H2 chimeras between H2-Db and -Dk, we demonstrate that the H2-Dk peptide-binding platform is required for Ly49P recognition. We identified m04 as a viral component necessary for recognition using a panel of MCMV-deletion mutant viruses and complementation of m04-deletion mutant (Δm04) virus infection. MA/My mice, which express Ly49P and H2-Dk, are resistant to MCMV; however, infection with Δm04 MCMV abrogates resistance. Depletion of NK cells in MA/My mice abrogates their resistance to wild-type MCMV infection, but does not significantly affect viral titers in mice infected with Δm04 virus, implicating NK cells in host protection through m04-dependent recognition. These findings reveal a novel mechanism of major histocompatability complex class I–restricted recognition of virally infected cells by an activating NK cell receptor.
Collapse
|
36
|
Miller-Kittrell M, Sparer TE. Feeling manipulated: cytomegalovirus immune manipulation. Virol J 2009; 6:4. [PMID: 19134204 PMCID: PMC2636769 DOI: 10.1186/1743-422x-6-4] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Accepted: 01/09/2009] [Indexed: 02/03/2023] Open
Abstract
No one likes to feel like they have been manipulated, but in the case of cytomegalovirus (CMV) immune manipulation, we do not really have much choice. Whether you call it CMV immune modulation, manipulation, or evasion, the bottom line is that CMV alters the immune response in such a way to allow the establishment of latency with lifelong shedding. With millions of years of coevolution within their hosts, CMVs, like other herpesviruses, encode numerous proteins that can broadly influence the magnitude and quality of both innate and adaptive immune responses. These viral proteins include both homologues of host proteins, such as MHC class I or chemokine homologues, and proteins with little similarity to any other known proteins, such as the chemokine binding protein. Although a strong immune response is launched against CMV, these virally encoded proteins can interfere with the host's ability to efficiently recognize and clear virus, while others induce or alter specific immune responses to benefit viral replication or spread within the host. Modulation of host immunity allows survival of both the virus and the host. One way of describing it would be a kind of "mutually assured survival" (as opposed to MAD, Mutually Assured Destruction). Evaluation of this relationship provides important insights into the life cycle of CMV as well as a greater understanding of the complexity of the immune response to pathogens in general.
Collapse
Affiliation(s)
- Mindy Miller-Kittrell
- Department of Microbiology, University of Tennessee, 1414 Cumberland Ave, Knoxville, TN, USA.
| | | |
Collapse
|
37
|
Cheeran MCJ, Jiang Z, Hu S, Ni HT, Palmquist JM, Lokensgard JR. Cytomegalovirus infection and interferon-gamma modulate major histocompatibility complex class I expression on neural stem cells. J Neurovirol 2008; 14:437-47. [PMID: 18937121 DOI: 10.1080/13550280802356845] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Cytomegalovirus (CMV) is the leading transmittable cause of congenital brain abnormalities in children and infection results in fatal ventriculoencephalitis in advanced acquired immunodeficiency syndrome (AIDS) patients. Pathology associated with CMV brain infection is seen predominantly in the periventricular region, an area known to harbor neural stem cells (NSCs). In the present study, using an adult model of murine CMV brain infection, the authors demonstrated that nestin-positive NSCs in the subventricular zone are susceptible to murine CMV infection. Furthermore, primary NSC cultures supported productive murine CMV replication with a 1000-fold increase in viral titers by 5 days post infection (d.p.i). Previous studies from the authors' laboratory demonstrated that CD8 lymphocytes were essential in protecting the brain against murine CMV infection. In the present study, the authors found that interferon (IFN)-gamma treatment increased the expression of major histocompatibility complex (MHC) class I on NSCs. Viral infection, on the other hand, inhibited this IFN-gamma-induced MHC up-regulation. In addition to increasing MHC class I expression, IFN-gamma (but not tumor necrosis factor [TNF]-alpha, interleukin [IL]-1 beta, or IL-10) also suppressed NSC proliferation in vitro. This decrease in proliferation was not accompanied by apoptosis or extracellular release of cellular lactate dehydrogenase (LDH), suggesting that the effects were not due to direct cytotoxicity. These studies demonstrate that NSCs are susceptible to murine CMV infection and inflammatory mediators, such as IFN-gamma, alter cellular characteristics which may have an impact on their reparative functions.
Collapse
Affiliation(s)
- Maxim C-J Cheeran
- Neuroimmunology Laboratory, Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Powers CJ, Früh K. Signal peptide-dependent inhibition of MHC class I heavy chain translation by rhesus cytomegalovirus. PLoS Pathog 2008; 4:e1000150. [PMID: 18833297 PMCID: PMC2542416 DOI: 10.1371/journal.ppat.1000150] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Accepted: 08/12/2008] [Indexed: 11/22/2022] Open
Abstract
The US2-11 region of human and rhesus cytomegalovirus encodes a conserved family of glycoproteins that inhibit MHC-I assembly with viral peptides, thus preventing cytotoxic T cell recognition. Since HCMV lacking US2-11 is no longer able to block assembly and transport of MHC-I, we examined whether this is also observed for RhCMV lacking the corresponding region. Unexpectedly, recombinant RhCMV lacking US2-11 was still able to inhibit MHC-I expression in infected fibroblasts, suggesting the presence of an additional MHC-I evasion mechanism. Progressive deletion analysis of RhCMV-specific genomic regions revealed that MHC-I expression is fully restored upon additional deletion of rh178. The protein encoded by this RhCMV-specific open reading frame is anchored in the endoplasmic reticulum membrane. In the presence of rh178, RhCMV prevented MHC-I heavy chain (HC) expression, but did not inhibit mRNA transcription or association of HC mRNA with translating ribosomes. Proteasome inhibitors stabilized a HC degradation intermediate in the absence of rh178, but not in its presence, suggesting that rh178 prevents completion of HC translation. This interference was signal sequence-dependent since replacing the signal peptide with that of CD4 or murine HC rendered human HCs resistant to rh178. We have identified an inhibitor of antigen presentation encoded by rhesus cytomegalovirus unique in both its lack of homology to any other known protein and in its mechanism of action. By preventing signal sequence-dependent HC translocation, rh178 acts prior to US2, US3 and US11 which attack MHC-I proteins after protein synthesis is completed. Rh178 is the first viral protein known to interfere at this step of the MHC-I pathway, thus taking advantage of the conserved nature of HC leader peptides, and represents a new mechanism of translational interference. To avoid immune detection by cytotoxic T lymphocytes, viruses interfere with antigen presentation by major histocompatibility complex class I (MHC-I) molecules. We have discovered a unique cytomegaloviral protein that interferes with the biosynthesis of MHC-I heavy chains and was thus termed viral inhibitor of heavy chain expression (VIHCE). We show that VIHCE does not affect transcription of MHC-I mRNA or the formation of poly-ribosomes. Surprisingly, however, very little MHC-I protein is detected, even when proteasomal protein degradation is inhibited, suggesting incomplete protein translation. Interestingly, VIHCE requires the proper MHC-I signal peptide, suggesting that CMV takes advantage of the high conservation of MHC-I signal peptides and interferes with protein translation by inhibiting signal sequence-dependent protein translocation. This is the first description of a viral protein that specifically targets the translation of a cellular immuno-stimulatory protein.
Collapse
Affiliation(s)
- Colin J. Powers
- Oregon Health and Science University, Vaccine and Gene Therapy Institute, Beaverton, Oregon, United States of America
| | - Klaus Früh
- Oregon Health and Science University, Vaccine and Gene Therapy Institute, Beaverton, Oregon, United States of America
- * E-mail:
| |
Collapse
|
39
|
Mouse cytomegalovirus microRNAs dominate the cellular small RNA profile during lytic infection and show features of posttranscriptional regulation. J Virol 2007; 81:13771-82. [PMID: 17942535 DOI: 10.1128/jvi.01313-07] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
MicroRNAs (miRNAs) are small, noncoding RNA molecules that regulate gene expression at the posttranscriptional level. Originally identified in a variety of organisms ranging from plants to mammals, miRNAs have recently been identified in several viruses. Viral miRNAs may play a role in modulating both viral and host gene expression. Here, we report on the identification and characterization of 18 viral miRNAs from mouse fibroblasts lytically infected with the murine cytomegalovirus (MCMV). The MCMV miRNAs are expressed at early times of infection and are scattered in small clusters throughout the genome with up to four distinct miRNAs processed from a single transcript. No significant homologies to human CMV-encoded miRNAs were found. Remarkably, as soon as 24 h after infection, MCMV miRNAs constituted about 35% of the total miRNA pool, and at 72 h postinfection, this proportion was increased to more than 60%. However, despite the abundance of viral miRNAs during the early phase of infection, the expression of some MCMV miRNAs appeared to be regulated. Hence, for three miRNAs we observed polyuridylation of their 3' end, coupled to subsequent degradation. Individual knockout mutants of two of the most abundant MCMV miRNAs, miR-m01-4 and miR-M44-1, or a double knockout mutant of miR-m21-1 and miR-M23-2, incurred no or only a very mild growth deficit in murine embryonic fibroblasts in vitro.
Collapse
|
40
|
Hassan J, Connell J. Translational mini-review series on infectious disease: congenital cytomegalovirus infection: 50 years on. Clin Exp Immunol 2007; 149:205-10. [PMID: 17635529 PMCID: PMC1941944 DOI: 10.1111/j.1365-2249.2007.03454.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cytomegalovirus (CMV) is the leading cause of congenital viral infection, with an incidence of 0.5-3% of live births worldwide. Clinical evidence has shown hearing and vision loss, mental retardation and sometimes death in affected newborns. Primary maternal CMV infection during gestation poses a 40% risk of intrauterine transmission in contrast to recurrent infection. European laboratories have made significant progress in the last decade in solving diagnostic problems linked to infection in pregnancy. With the advances in CMV serology, such as detection of anti-CMV IgM by enzyme immunoassays (EIA), confirmed by Western blot, together with seroconversion and anti-CMV IgG avidity evaluation in pregnant mothers, can help to identify recent infection. Preventative measures such as screening for CMV in the routine serological work-up of pregnant women have been introduced in countries such as Spain and Italy. The development of specific T cell-mediated immune responses in mothers, fetus and neonates is now emerging with regard to antigen-specific CD4 and CD8 T cells, differentiation status, proliferative and cytokine responses. A protective vaccine against CMV is a major public health priority and the study of vaccines in animal model systems has identified potential strategies for interrupting transmission and preventing disease in newborns. Congenital CMV infection has a variable outcome and therefore novel diagnostic methods are required to identify those at risk and therapeutic interventions are needed to improve the long-term prognosis of those infected. CMV was first isolated in 1957. We are now 50 years on, so procrastination is not an option.
Collapse
Affiliation(s)
- J Hassan
- National Virus Reference Laboratory and Centre for Research into Infectious Disease, University College Dublin, Dublin, Ireland.
| | | |
Collapse
|
41
|
Corbett AJ, Forbes CA, Moro D, Scalzo AA. Extensive sequence variation exists among isolates of murine cytomegalovirus within members of the m02 family of genes. J Gen Virol 2007; 88:758-769. [PMID: 17325348 DOI: 10.1099/vir.0.82623-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Murine cytomegalovirus (MCMV) is a widely used model for human cytomegalovirus (HCMV) and has facilitated many important discoveries about the biology of CMVs. Most of these studies are conducted using the laboratory MCMV strains Smith and K181. However, wild-derived isolates of MCMV, like HCMV clinical isolates, exhibit genetic variation from laboratory strains, particularly at the ends of their genomes in areas containing known or putative immune-evasion and tropism genes. This study analysed the nucleotide sequence of the m02-m05 region, within the m02 gene family, of a number of laboratory and wild-derived MCMV isolates, and found a large degree of variation in both the sequence and arrangement of genes. A new open reading frame (ORF), designated m03.5, was found to be present in a number of wild isolates of MCMV in place of m03. Two distinct isolates, W8 and W8211, were found to possess both m03 and m03.5. Both m03 and m03.5 had early transcription kinetics and the encoded proteins could be detected on the cell surface, consistent with a possible role in immune evasion through binding to host-cell proteins. These data show that gene duplication and sequence variation occur within different isolates of MCMV found in the wild. As this variation among strains may alter the function of genes, these findings should be considered when analysing gene function or host-virus interactions in laboratory models.
Collapse
Affiliation(s)
- Alexandra J Corbett
- Centre for Experimental Immunology, Lions Eye Institute, 2 Verdun Street, Nedlands, WA 6009, Australia
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, University of Western Australia, Nedlands, WA 6009, Australia
| | - Catherine A Forbes
- Centre for Experimental Immunology, Lions Eye Institute, 2 Verdun Street, Nedlands, WA 6009, Australia
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, University of Western Australia, Nedlands, WA 6009, Australia
| | - Dorian Moro
- School of Natural Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Anthony A Scalzo
- Centre for Experimental Immunology, Lions Eye Institute, 2 Verdun Street, Nedlands, WA 6009, Australia
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, University of Western Australia, Nedlands, WA 6009, Australia
| |
Collapse
|
42
|
Hess CM, Wang Z, Edwards SV. Evolutionary genetics of Carpodacus mexicanus, a recently colonized host of a bacterial pathogen, Mycoplasma gallisepticum. Genetica 2006; 129:217-25. [PMID: 17103248 DOI: 10.1007/s10709-006-9016-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2005] [Accepted: 02/28/2006] [Indexed: 10/23/2022]
Abstract
We present molecular data documenting how introduction to the eastern United States and an epizootic involving a bacterial pathogen has affected the genetic diversity of house finches, a cardueline songbird. Population bottlenecks during introduction can cause loss of genetic variation and may negatively affect a population's ability to adapt to novel stressors such as disease. Although a genome-wide survey using Amplified Fragment Length Polymorphism (AFLP) markers suggests little loss of genetic diversity in introduced populations, an epizootic of bacterial Mycoplasma has nonetheless caused dramatic declines in the eastern US population. Sequence analysis of a candidate gene for pathogen resistance in the Major Histocompatibity Complex (MHC) in pre- and post-epizootic population samples reveals allele frequency shifts since introduction of the pathogen, but similar shifts are also observed in control populations not exposed to the bacteria, and in a neutral non-coding locus. Expression studies using a novel subtractive hybridization approach indicate decreased expression of the class II MHC locus upon exposure to Mycoplasma, a pattern also seen in MHC class I loci in mice infected with cytomegalovirus and consistent with manipulation of the finch immune system by Mycoplasma. These results will be further expanded using experimental studies as well as examination of evolution of the pathogen genome itself.
Collapse
|
43
|
Pinto AK, Munks MW, Koszinowski UH, Hill AB. Coordinated function of murine cytomegalovirus genes completely inhibits CTL lysis. THE JOURNAL OF IMMUNOLOGY 2006; 177:3225-34. [PMID: 16920962 DOI: 10.4049/jimmunol.177.5.3225] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Murine CMV (MCMV) encodes three viral genes that interfere with Ag presentation (VIPRs) to CD8 T cells, m04, m06, and m152. Because the functional impact of these genes during normal infection of C57BL/6 mice is surprisingly modest, we wanted to determine whether the VIPRs are equally effective against the entire spectrum of H-2(b)-restricted CD8 T cell epitopes. We also wanted to understand how the VIPRs interact at a functional level. To address these questions, we used a panel of MCMV mutants lacking each VIPR in all possible combinations, and CTL specific for 15 H-2(b)-restricted MCMV epitopes. Only expression of all three MCMV VIPRs completely inhibited killing by CTL specific for all 15 epitopes, but removal of any one VIPR enabled lysis by at least some CTL. The dominant interaction between the VIPRs was cooperation: m06 increased the inhibition of lysis achieved by either m152 or m04. However, for 1 of 15 epitopes m04 functionally antagonized m152. There was little differential impact of any of the VIPRs on K(b) vs D(b), but a surprising degree of differential impact of the three VIPRs for different epitopes. These epitope-specific differences did not correlate with functional avidity, or with timing of VIPR expression in relation to Ag expression in the virus replication cycle. Although questions remain about the molecular mechanism and in vivo role of these genes, we conclude that the coordinated function of MCMV's three VIPRs results in a powerful inhibition of lysis of infected cells by CD8 T cells.
Collapse
Affiliation(s)
- Amelia K Pinto
- Oregon Health and Science University, Molecular Microbiology and Immunology, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
44
|
Andoniou CE, Degli-Esposti MA. Insights into the mechanisms of CMV‐mediated interference with cellular apoptosis. Immunol Cell Biol 2006; 84:99-106. [PMID: 16405657 DOI: 10.1111/j.1440-1711.2005.01412.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Apoptosis has the potential to function as a defence mechanism during viral infection. Identification of CMV mutants that cause the apoptotic death of infected cells confirmed that viral infection activates apoptotic pathways and that this process is counteracted by CMV to ensure efficient viral replication. The recent identification of CMV-encoded proteins that suppress cell death has greatly enhanced our understanding of the mechanisms used by this family of viruses to prevent apoptosis. CMV do not encode homologues of known death-suppressing proteins, suggesting that the CMV family has evolved novel, more sophisticated strategies for the inhibition of apoptosis. The identification and characterization of the human CMV (HCMV)-encoded antiapoptotic proteins UL36 (viral inhibitor of caspase-8 activation [vICA]) and UL37 (viral mitochondria-localized inhibitor of apoptosis [vMIA]) have confirmed that CMV target unique apoptotic control points. For example, vMIA inhibits apoptosis by binding Bax and sequestering it at the mitochondrial membrane as an inactive oligomer. This knowledge not only provides a more complete understanding of the CMV replication process but also allows the identification of previously unrecognized apoptotic checkpoints. Because HCMV is an important cause of birth defects and an increasingly important opportunistic pathogen, a firm grasp of the mechanisms by which it affects cellular apoptosis may provide avenues for the design of improved therapeutic strategies. Here, we review the recent progress made in understanding the role of CMV-encoded proteins in the inhibition of apoptosis.
Collapse
Affiliation(s)
- Christopher E Andoniou
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
| | | |
Collapse
|
45
|
Abstract
Relatively small genomes and high replication rates allow viruses and bacteria to accumulate mutations. This continuously presents the host immune system with new challenges. On the other side of the trenches, an increasingly well-adjusted host immune response, shaped by coevolutionary history, makes a pathogen's life a rather complicated endeavor. It is, therefore, no surprise that pathogens either escape detection or modulate the host immune response, often by redirecting normal cellular pathways to their advantage. For the purpose of this chapter, we focus mainly on the manipulation of the class I and class II major histocompatibility complex (MHC) antigen presentation pathways and the ubiquitin (Ub)-proteasome system by both viral and bacterial pathogens. First, we describe the general features of antigen presentation pathways and the Ub-proteasome system and then address how they are manipulated by pathogens. We discuss the many human cytomegalovirus (HCMV)-encoded immunomodulatory genes that interfere with antigen presentation (immunoevasins) and focus on the HCMV immunoevasins US2 and US11, which induce the degradation of class I MHC heavy chains by the proteasome by catalyzing their export from the endoplasmic reticulum (ER)-membrane into the cytosol, a process termed ER dislocation. US2- and US11-mediated subversion of ER dislocation ensures proteasomal degradation of class I MHC molecules and presumably allows HCMV to avoid recognition by cytotoxic T cells, whilst providing insight into general aspects of ER-associated degradation (ERAD) which is used by eukaryotic cells to purge their ER of defective proteins. We discuss the similarities and differences between the distinct pathways co-opted by US2 and US11 for dislocation and degradation of human class I MHC molecules and also a putatively distinct pathway utilized by the murine herpes virus (MHV)-68 mK3 immunoevasin for ER dislocation of murine class I MHC. We speculate on the implications of the three pathogen-exploited dislocation pathways to cellular ER quality control. Moreover, we discuss the ubiquitin (Ub)-proteasome system and its position at the core of antigen presentation as proteolysis and intracellular trafficking rely heavily on Ub-dependent processes. We add a few examples of manipulation of the Ub-proteasome system by pathogens in the context of the immune system and such diverse aspects of the host-pathogen relationship as virus budding, bacterial chromosome integration, and programmed cell death, to name a few. Finally, we speculate on newly found pathogen-encoded deubiquitinating enzymes (DUBs) and their putative roles in modulation of host-pathogen interactions.
Collapse
Affiliation(s)
- Joana Loureiro
- Whitehead Institute, 9 Cambridge Center, Cambridge, Massachusetts, USA
| | | |
Collapse
|
46
|
Gumá M, Angulo A, López-Botet M. NK cell receptors involved in the response to human cytomegalovirus infection. Curr Top Microbiol Immunol 2005; 298:207-23. [PMID: 16323417 DOI: 10.1007/3-540-27743-9_11] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human cytomegalovirus (HCMV) infection is a paradigm of the complexity reached by host-pathogen interactions. To avoid recognition by cytotoxic T lymphocytes (CTL) HCMV inhibits the expression of HLA class I molecules. As a consequence, engagement of inhibitory killer immunoglobulin-like receptors (KIR), CD94/NKG2A, and CD85j (ILT2 or LIR-1) natural killer cell receptors (NKR) specific for HLA class I molecules is impaired, and infected cells become vulnerable to an NK cell response driven by activating receptors. In addition to the well-defined role of the NKG2D lectin-like molecule, the involvement of other triggering receptors (i.e., activating KIR, CD94/NKG2C, NKp46, NKp44, and NKp30) in the response to HCMV is being explored. To escape from NK cell-mediated surveillance, HCMV interferes with the expression of NKG2D ligands in infected cells. In addition, the virus may keep NK inhibitory receptors engaged preserving HLA class I molecules with a limited role in antigen presentation (i.e., HLA-E) or, alternatively, displaying class I surrogates. Despite considerable progress in the field, a number of issues regarding the involvement of NKR in the innate immune response to HCMV remain uncertain.
Collapse
Affiliation(s)
- M Gumá
- Molecular Immunopathology Unit, DCEXS, Universitat Pompeu Fabra, Dr. Aiguader 80, 08003 Barcelona, Spain
| | | | | |
Collapse
|
47
|
Rich RF, Cook WJ, Green WR. Spontaneous in vivo retrovirus-infected T and B cells, but not dendritic cells, mediate antigen-specific Fas ligand/Fas-dependent apoptosis of anti-retroviral CTL. Virology 2005; 346:287-300. [PMID: 16337984 DOI: 10.1016/j.virol.2005.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2005] [Revised: 07/14/2005] [Accepted: 10/12/2005] [Indexed: 12/14/2022]
Abstract
C57BL/6 (H-2b), but not spontaneous virus-expressing AKR.H-2b congenic, mice generate retrovirus-specific CD8+ CTL responses to the immunodominant Kb-restricted epitope, KSPWFTTL. AKR.H-2b non-responsiveness is mediated by a peripheral tolerance mechanism. When co-cultured with primed B6 antiviral pCTL, AKR.H-2b splenocytes are recognized by the antiviral TcR as "veto" cells, which inhibit by an exquisitely virus-specific, MHC-restricted, veto cell FasL/responder T cell Fas, mediated apoptotic mechanism. Here, AKR.H-2b thymus, lymph node, and bone marrow cells are also shown to inhibit antiviral CTL generation. Purified AKR.H-2b CD4+ and CD8+ T cells, and B cells, served effectively as FasL-dependent veto cells. In contrast, AKR.H-2b dendritic cells (DC) did not efficiently veto antiviral CTL responses, despite expressing sufficient MHC class I/viral peptide complexes for TcR recognition. AKR.H-2b DC also expressed FasL mRNA and cell surface protein, albeit at a lower level than AKR.H-2b T and B cells. These findings suggest a fail-safe escape mechanism by virus-infected cells for escape from CTL-mediated immunity.
Collapse
Affiliation(s)
- Robert F Rich
- Department of Microbiology and Immunology and the Norris Cotton Cancer Center, Dartmouth Medical School, 1 Medical Center Drive, Borwell 603 West, Lebanon, New Hampshire 03756, USA
| | | | | |
Collapse
|
48
|
Hassink GC, Duijvestijn-van Dam JG, Koppers-Lalic D, van Gaans-van den Brink J, van Leeuwen D, Vink C, Bruggeman CA, Wiertz EJHJ. Rat Cytomegalovirus Induces a Temporal Downregulation of Major Histocompatibility Complex Class I Cell Surface Expression. Viral Immunol 2005; 18:607-15. [PMID: 16359227 DOI: 10.1089/vim.2005.18.607] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Herpesviruses are known to influence expression of major histocompatibility complex (MHC) class I molecules on the surface of infected cells using a variety of mechanisms. Downregulation of MHC class I expression prohibits detection and elimination of infected cells by cytotoxic T lymphocytes. To investigate the effect of rat cytomegalovirus (RCMV) infection on MHC class I expression, we infected immortalized and primary rat fibroblasts with RCMV and monitored surface expression of MHC class I molecules at various time-points postinfection. These experiments revealed a downregulation of MHC class I surface expression by RCMV, a phenomenon that has also been reported for human and murine CMV. However, in contrast to the other cytomegaloviruses, RCMV causes only a temporal downregulation of MHC class I, with a maximal decrease at 12 h postinfection. Unlike murine and human CMV, RCMV does not induce proteolytic degradation of MHC class I molecules. In RCMV-infected cells, the MHC class I molecules are stable, but their exit from the ER is delayed.
Collapse
Affiliation(s)
- Gerco C Hassink
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Wang Z, Farmer K, Hill GE, Edwards SV. A cDNA macroarray approach to parasite-induced gene expression changes in a songbird host: genetic response of house finches to experimental infection by Mycoplasma gallisepticum. Mol Ecol 2005; 15:1263-73. [PMID: 16626453 DOI: 10.1111/j.1365-294x.2005.02753.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In 1994, the bacterial parasite Mycoplasma gallisepticum expanded its host range and swept through populations of a novel host--eastern US populations of the house finch (Carpodacus mexicanus). This epizootic caused a dramatic decline in finch population numbers, has been shown to have caused strong selection on house finch morphology, and presumably caused evolutionary change at the molecular level as finches evolved enhanced resistance. As a first step toward identifying finch genes that respond to infection by Mycoplasma and which may have experienced natural selection by this parasite, we used suppression subtractive hybridization (SSH) and cDNA macroarray approaches to identify differentially expressed genes regulated by the Mycoplasma parasite. Two subtractive cDNA libraries consisting of 16,512 clones were developed from spleen using an experimentally uninfected bird as the 'tester' and an infected bird as 'driver', and vice versa. Two hundred and twenty cDNA clones corresponding 34 genes with known vertebrate homologues and a large number of novel transcripts were found to be qualitatively up- or down-regulated genes by high-density filter hybridization. These gene expression changes were further confirmed by a high throughout reverse Northern blot approach and in specific cases by targeted Northern analysis. blast searches show that heat shock protein (HSP) 90, MHC II-associated invariant chain (CD74), T-cell immunoglobulin mucin 1 (TIM1), as well as numerous novel expressed genes not found in the databases were up- or down-regulated by the host in response to this parasite. Our results and macroarray resources provide a foundation for molecular co-evolutionary studies of the Mycoplasma parasite and its recently colonized avian host.
Collapse
Affiliation(s)
- Zhenshan Wang
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | | | | | | |
Collapse
|
50
|
Pinto AK, Hill AB. Viral Interference with Antigen Presentation to CD8+T Cells: Lessons from Cytomegalovirus. Viral Immunol 2005; 18:434-44. [PMID: 16212522 DOI: 10.1089/vim.2005.18.434] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cytomegaloviruses (CMV), in common with other Herpesviruses, establish lifelong persistence in their hosts. These highly host-specific viruses each encode viral genes that interfere with antigen presentation to CD8+ T cells, although the molecular mechanisms by which this end is achieved differ for human and murine CMVs. In each case, there has been a presumption that these genes are necessary for virus persistence in the host. However, recent data in the murine model casts doubt on that presumption. Here, we review the molecular mechanisms of interference with the class I pathway, and the published data regarding functional significance, with a focus on the murine model.
Collapse
Affiliation(s)
- Amelia K Pinto
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | | |
Collapse
|