1
|
Zhang X, Wang Q, Han S, Song G, Wang B, Wang Y. Human cytomegalovirus-IE2 suppresses antigen presentation of macrophage through the IL10/STAT3 signalling pathway in transgenic mouse. PLoS One 2025; 20:e0322334. [PMID: 40323903 PMCID: PMC12052161 DOI: 10.1371/journal.pone.0322334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/20/2025] [Indexed: 05/07/2025] Open
Abstract
Human cytomegalovirus (HCMV) has evolved sophisticated strategies to evade host immune defenses, enabling its persistent survival in human populations. HCMV intermediate-early protein 2 (IE2) has been identified as a crucial factor in immune evasion mechanisms. However, the specific immunomodulatory effects of IE2 on antigen presentation remain insufficiently explored. In this study, we established a transgenic mouse model to systematically examine the impact and molecular mechanisms of IE2 on macrophages (Mφs) antigen presentation in vivo. Our findings demonstrated that IE2 modifies Mφs' function by preventing their phagocytic activity and polarization. Additionally, IE2 inhibits Mφs overactivation both in vivo and in vitro, which raises IL-10 levels and activates the downstream mediator STAT3, which in turn decreases T cell immune responses by encouraging T helper 2 (Th2) type responses. In conclusion, these findings underscore the potential of IE2 as a critical regulator of immune evasion and may contribute to the development of novel, targeted therapeutic strategies against the virus.
Collapse
Affiliation(s)
- Xianjuan Zhang
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qing Wang
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shuo Han
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guanghui Song
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Wang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yunyang Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Penner I, Krämer N, Hirsch J, Büscher N, Schmidt H, Plachter B. Deletion of the Human Cytomegalovirus US2 to US11 Gene Family Members Impairs the Type-I Interferon Response. Viruses 2025; 17:426. [PMID: 40143353 PMCID: PMC11945591 DOI: 10.3390/v17030426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/07/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Infection of cells with the human cytomegalovirus (HCMV) triggers the expression of interferon-stimulated genes (ISGs). ISGs encode proteins with antiviral functions, such as inhibiting viral replication, promoting cell death of infected cells and enhancing immune responses. HCMV has evolved mechanisms to evade the antiviral effects of ISGs. The viral proteins encoded by the viral genes US7, US8, and US9 have been shown to interfere with interferon induction. US7 to US9 are embedded in a cluster of HCMV genes, termed US2 to US11. The individual members of this gene family interfere on multiple levels with innate and adaptive immune responses to HCMV infection. Using viral mutants with different deletions in US2 to US11, we addressed the question if genes other than US7 to US9 would also influence the IFN responses. Surprisingly, deletion of the complete US2 to US11 gene region led to reduced levels of selected ISGs. Cells infected with viruses in which individual US2 to US11 genes were deleted showed a less pronounced reduction of the selected ISGs. The experiments including RNA-seq analyses indicate that genes of the US2 to US11 gene family have a complex interaction with the IFN-ISG response which is likely regulated on the level of ISG protein stability. As US2-US11 are dispensable for replication in cell culture, the genomic region was frequently used for the insertion of bacterial artificial chromosome vectors in the process of cloning the complete HCMV genome. The results shown here must be considered when viruses derived from BACs with US2-US11 deletions are used and whether appropriate controls must be applied.
Collapse
|
3
|
Blanco R, Muñoz JP. Molecular Insights into HR-HPV and HCMV Co-Presence in Cervical Cancer Development. Cancers (Basel) 2025; 17:582. [PMID: 40002177 PMCID: PMC11853276 DOI: 10.3390/cancers17040582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/27/2024] [Accepted: 11/03/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Cervical cancer remains a significant health concern worldwide and the primary cause of cancerous cervical lesions is the infection with high-risk human papillomavirus (HR-HPV). However, emerging evidence suggests that HR-HPV infection alone is insufficient for cancer development, and other co-factors may contribute to cervical carcinogenesis. Human cytomegalovirus (HCMV), a common herpesvirus frequently detected in cervical cancer samples, has demonstrated oncogenic potential. OBJECTIVES This review aims to explore the molecular interactions between HR-HPV and HCMV in promoting cervical cancer progression. METHODS A comprehensive search was conducted in PubMed and Google Scholar, focusing on articles examining the role of HCMV in cervical tissues and/or cells, selected based on relevance and significance. RESULTS The reviewed literature indicates that HCMV and HR-HPV share several oncogenic mechanisms that could drive cervical cell transformation. CONCLUSIONS Both viruses may synergistically promote cervical epithelial transformation and tumor progression in multiple ways. HR-HPV may facilitate HCMV entry by increasing host cell receptors essential for viral attachment. Additionally, HR-HPV and HCMV may cooperatively disrupt cellular processes, enhancing carcinogenesis. Both viruses may also modulate the local immune environment, enabling immune evasion and lesion persistence. However, further in vitro and in vivo studies are required to validate these hypotheses.
Collapse
Affiliation(s)
- Rancés Blanco
- Independent Researcher, Av. Vicuña Mackenna Poniente 6315, La Florida 8240000, Chile
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| |
Collapse
|
4
|
Blanco R, Muñoz JP. Human Cytomegalovirus Infection and Breast Cancer: A Literature Review of Clinical and Experimental Data. BIOLOGY 2025; 14:174. [PMID: 40001942 PMCID: PMC11851556 DOI: 10.3390/biology14020174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 02/27/2025]
Abstract
Breast cancer (BC) remains a significant global health challenge, highlighting the need for continued research into novel risk factors, diagnostic approaches, and personalized treatments. Among emerging risk factors, viral infections have been implicated as potential contributors to breast carcinogenesis and BC progression. Recent evidence suggests that specific oncogenic strains of human cytomegalovirus (HCMV) may have the capacity to transform human mammary epithelial cells. This review assesses clinical data regarding HCMV presence in both tumor and non-tumor breast tissues, examining the role of HCMV oncoproteins in BC development and progression. Current findings indicate a higher prevalence of HCMV infection in breast carcinomas compared to non-tumor tissues, associated with an elevated risk of BC. Additionally, the HCMV-driven breast carcinogenesis model proposed here suggests that HCMV oncoproteins may activate multiple oncogenic pathways, fostering cell proliferation, survival, and tumor development. A deeper understanding of the role of HCMV in BC could enhance risk stratification and support the creation of targeted therapeutic strategies.
Collapse
Affiliation(s)
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| |
Collapse
|
5
|
Okpoluaefe S, Ismail IS, Mohamed R, Hassan N. Adaptive natural killer cell expression in response to cytomegalovirus infection in blood and solid cancer. Heliyon 2024; 10:e32622. [PMID: 38961938 PMCID: PMC11219991 DOI: 10.1016/j.heliyon.2024.e32622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024] Open
Abstract
Natural Killer (NK) cells are conventionally thought to be an indefinite part of innate immunity. However, in a specific subset of NK cells, recent data signify an extension of their "duties" in immune surveillance and response, having characteristics of adaptive immunity, in terms of persistence and cytotoxicity. These cells are known as the adaptive or memory-like NK cells, where human cytomegalovirus (HCMV) infection has been shown to drive the expansion of adaptive NKG2C+ NK cells. HCMV is a ubiquitous pathogen whose prevalence differs worldwide with respect to the socioeconomic status of countries. The adaptive NK cell subpopulation is often characterized by the upregulated expression of NKG2C, CD16, and CD2, and restricted expression of NKG2A, FCεRγ and killer immunoglobulin-like receptors (KIR), although these phenotypes may differ in different disease groups. The reconfiguration of these receptor distributions has been linked to epigenetic factors. Hence, this review attempts to appraise literature reporting markers associated with adaptive or memory-like NK cells post-HCMV infection, in relation to solid cancers and hematological malignancies. Adaptive NK cells, isolated and subjected to ex vivo modifications, have the potential to enhance anti-tumor response which can be a promising strategy for adoptive immunotherapy.
Collapse
Affiliation(s)
- Suruthimitra Okpoluaefe
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Bertam, Kepala Batas, Penang, Malaysia
- Emerging Infectious Disease Group, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 3200 Bertam, Kepala Batas, Penang, Malaysia
| | - Ida Shazrina Ismail
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Bertam, Kepala Batas, Penang, Malaysia
- Breast Cancer Translational Research Program, BCTRP@IPPT, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Bertam, Kepala Batas, Penang, Malaysia
| | - Rafeezul Mohamed
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Bertam, Kepala Batas, Penang, Malaysia
- Breast Cancer Translational Research Program, BCTRP@IPPT, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Bertam, Kepala Batas, Penang, Malaysia
| | - Norfarazieda Hassan
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Bertam, Kepala Batas, Penang, Malaysia
- Breast Cancer Translational Research Program, BCTRP@IPPT, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Bertam, Kepala Batas, Penang, Malaysia
- Emerging Infectious Disease Group, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 3200 Bertam, Kepala Batas, Penang, Malaysia
| |
Collapse
|
6
|
Wang H, Peng W, Wang J, Zhang C, Zhao W, Ran Y, Yang X, Chen J, Li H. Human Cytomegalovirus UL23 Antagonizes the Antiviral Effect of Interferon-γ by Restraining the Expression of Specific IFN-Stimulated Genes. Viruses 2023; 15:v15041014. [PMID: 37112994 PMCID: PMC10145438 DOI: 10.3390/v15041014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Interferon-γ (IFN-γ) is a critical component of innate immune responses in humans to combat infection by many viruses, including human cytomegalovirus (HCMV). IFN-γ exerts its biological effects by inducing hundreds of IFN-stimulated genes (ISGs). In this study, RNA-seq analyses revealed that HCMV tegument protein UL23 could regulate the expression of many ISGs under IFN-γ treatment or HCMV infection. We further confirmed that among these IFN-γ stimulated genes, individual APOL1 (Apolipoprotein-L1), CMPK2 (Cytidine/uridine monophosphate kinase 2), and LGALS9 (Galectin-9) could inhibit HCMV replication. Moreover, these three proteins exhibited a synergistic effect on HCMV replication. UL23-deficient HCMV mutants induced higher expression of APOL1, CMPK2, and LGALS9, and exhibited lower viral titers in IFN-γ treated cells compared with parental viruses expressing full functional UL23. Thus, UL23 appears to resist the antiviral effect of IFN-γ by downregulating the expression of APOL1, CMPK2, and LGALS9. This study highlights the roles of HCMV UL23 in facilitating viral immune escape from IFN-γ responses by specifically downregulating these ISGs.
Collapse
Affiliation(s)
- Hankun Wang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Weijian Peng
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jialin Wang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Chunling Zhang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Wangchun Zhao
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yanhong Ran
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xiaoping Yang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jun Chen
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Guangzhou 510632, China
| | - Hongjian Li
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Guangzhou 510632, China
| |
Collapse
|
7
|
Mosher BS, Kowalik TF, Yurochko AD. Overview of how HCMV manipulation of host cell intracellular trafficking networks can promote productive infection. FRONTIERS IN VIROLOGY 2022. [DOI: 10.3389/fviro.2022.1026452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human cytomegalovirus (HCMV) is a significant cause of morbidity and mortality in the immunocompromised and developing fetuses. Infection has also been linked to chronic inflammatory diseases, cardiovascular disease, and the development of certain cancers. The wide range of pathologies associated with HCMV infection is attributable to the broad cellular tropism of the virus where infection affects every organ system. Like other viruses, HCMV must tailor host cells to support productive infection. In particular, HCMV dedicates many resources and various strategies to manipulate host intracellular trafficking networks to facilitate various aspects of infection across all infected cell types. The dysregulation of host intracellular trafficking networks allows the virus to translocate to the host cell nucleus for genome replication, facilitate nuclear import/export of viral proteins and immature virions, subvert the host immune response, form new organelles for progeny virion assembly, maturation and egress, and promote cellular migration and viral spread. However, due to their complex nature, many aspects of these processes are not well-studied. New research and omics-based technologies have recently begun to elucidate the extent to which HCMV dysregulates host cell trafficking machinery. Here we review the variety of strategies HCMV utilizes to dysregulate intracellular trafficking networks to promote productive infection.
Collapse
|
8
|
Advances in DNA- and RNA-Based Oncolytic Viral Therapeutics and Immunotherapies. Appl Microbiol 2022. [DOI: 10.3390/applmicrobiol2020024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The role of viruses has been studied extensively for use as curative cancer therapies. However, the natural immunogenicity of viruses and their interaction with the host’s immune system needs to be examined to determine the full effectiveness of the viral treatment. The prevalence of cancer is increasing globally, and treatments are needed to support the increasing body of patient care. Oncolytic viral therapies used existing pathogenic viruses, which are genetically modified to not cause disease in humans when administered using a vaccine viral vector. Immunotherapies are another avenue of recent interest that has gained much traction. This review will discuss oncolytic viral approaches using three DNA-based viruses, including herpes simplex virus (HSV), vaccinia virus, and adenovirus; as well as four RNA-based viruses, including reovirus, Newcastle disease virus (NDV), poliovirus, and measles virus (MV). It also examines the novel field of cancer-based immunotherapies.
Collapse
|
9
|
Abstract
While many viral infections are limited and eventually resolved by the host immune response or by death of the host, other viruses establish long-term relationships with the host by way of a persistent infection, that range from chronic viruses that may be eventually cleared to those that establish life-long persistent or latent infection. Viruses infecting hosts from bacteria to humans establish quiescent infections that must be reactivated to produce progeny. For mammalian viruses, most notably herpesviruses, this quiescent maintenance of viral genomes in the absence of virus replication is referred to as latency. The latent strategy allows the virus to persist quiescently within a single host until conditions indicate a need to reactivate to reach a new host or, to re-seed a reservoir within the host. Here, I review common themes in viral strategies to regulate the latent cycle and reactivate from it ranging from bacteriophage to herpesviruses with a focus on human cytomegalovirus (HCMV). Themes central to herpesvirus latency include, epigenetic repression of viral gene expression and mechanisms to regulate host signaling and survival. Critical to the success of a latent program are mechanisms by which the virus can "sense" fluctuations in host biology (within the host) or environment (outside the host) and make appropriate "decisions" to maintain latency or re-initiate the replicative program. The signals or environments that indicate the establishment of a latent state, the very nature of the latent state, as well as the signals driving reactivation have been topics of intense study from bacteriophage to human viruses, as these questions encompass the height of complexity in virus-host interactions-where the host and the virus coexist.
Collapse
Affiliation(s)
- Felicia Goodrum
- Department of Immunobiology, BIO5 Institute, University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
10
|
Comprehensive Analysis of Human Cytomegalovirus- and HIV-Mediated Plasma Membrane Remodeling in Macrophages. mBio 2021; 12:e0177021. [PMID: 34399625 PMCID: PMC8406226 DOI: 10.1128/mbio.01770-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The plasma membrane (PM) must be overcome by viruses during entry and release. Furthermore, the PM represents the cellular communication compartment and the immune system interface. Hence, viruses have evolved sophisticated strategies to remodel the PM, for instance to avoid immune sensing and clearance of infected cells. We performed a comprehensive analysis of cell surface dysregulation by two human-pathogenic viruses, human cytomegalovirus (HCMV) and human immunodeficiency virus type 1 (HIV-1), in primary macrophages, which are classical antigen-presenting cells and orchestrators of the immune system. Scanning ion conductance microscopy revealed a loss of roughness and an overall smooth phenotype of HCMV-infected macrophages, in contrast to HIV-1 infection. This phenotype was also evident on the molecular level. When we screened for cell surface receptors modulated by HCMV, 42 of 332 receptors tested were up- or downregulated, whereas HIV-1 affected only 7 receptors. In particular CD164, CD84, and CD180 were targeted by HCMV. Mechanistically, HCMV induced transcriptional silencing of these receptors in an interferon (IFN)-independent manner, and expression was reduced not only by lab-adapted HCMV but also by clinical HCMV isolates. Altogether, our plasma membrane profiling of human macrophages provides clues to understand how viruses evade the immune system and identified novel cell surface receptors targeted by HCMV. IMPORTANCE The PM is a key component that viruses have to cope with. It is a barrier for infection and egress and is critically involved in antiviral immune signaling. We hence asked the question how two immunomodulatory viruses, HIV-1 and HCMV, dysregulate this compartment in infected macrophages, relevant in vivo targets of both viruses. We employed a contact-free microscopic technique to image the PM of infected cells and performed a phenotypic flow cytometry-based screen to identify receptor modulations on a molecular level. Our results show that HIV-1 and HCMV differentially manipulate the PM of macrophages. While HIV-1-mediated changes are relatively subtle, HCMV induces major alterations of the PM. We identify novel immune receptors manipulated by HCMV and define mechanisms of how HCMV interferes with receptor expression. Altogether, our study reveals differential strategies of how two human-pathogenic viruses manipulate infected cells and identifies potential novel pathways of HCMV immune evasion.
Collapse
|
11
|
Yoo SG, Han KD, Lee KH, Lim J, La Y, Kwon DE, Han SH. Epidemiological changes in cytomegalovirus end-organ diseases in a developed country: A nationwide, general-population-based study. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2021; 55:812-819. [PMID: 34475004 DOI: 10.1016/j.jmii.2021.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/29/2021] [Accepted: 08/05/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) can cause tissue-invasive diseases in various organs after primary infection or through reactivation of latent-to-lytic switch over a lifetime. The number of individuals who are at risk of CMV diseases, such as elderly or immunocompromised patients, is constantly increasing; however, recent epidemiological changes associated with CMV disease have not been fully evaluated. METHODS We used claims data of about 50 million individuals between 2010 and 2015 from the Korean Health Insurance Review and Assessment Service nationwide database. The code for CMV end-organ diseases in the 'Relieved Co-payment Policy' program matches the ICD-10 code of B25, except for congenital CMV infection and mononucleosis. A 628 cases of CMV and 3140 controls (without CMV disease), matched for age and sex, were selected from this dataset in order to evaluate the effect of adult CMV diseases on all-cause death. RESULTS The overall unadjusted incidence rate (IR) of CMV end-organ diseases was 0.52/100,000 individuals. The standardized IR, adjusted for age and sex, have continuously increased from 0.32/100,000 in 2010 to 0.75/100,000 in 2015. The overall unadjusted IR in adult population was highest in 70-79 years for six years (0.96/100,000). In the model adjusted for age, sex, immunocompromised status including solid-organ or hematopoietic stem cell transplant recipients, hematologic malignancies, and human immunodeficiency virus diseases, the hazard ratio of case group was 5.2 (95% confidence interval, 3.6-7.4) for all-cause mortality. CONCLUSION Nationwide data indicates that CMV end-organ disease has steadily increased in the past six years and is associated with higher mortality.
Collapse
Affiliation(s)
- Seul Gi Yoo
- Divison of Infectious Disease, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung Do Han
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyoung Hwa Lee
- Divison of Infectious Disease, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joohee Lim
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yeonju La
- Divison of Infectious Disease, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Da Eun Kwon
- Divison of Infectious Disease, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Hoon Han
- Divison of Infectious Disease, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Targeting the latent human cytomegalovirus reservoir for T-cell-mediated killing with virus-specific nanobodies. Nat Commun 2021; 12:4436. [PMID: 34290252 PMCID: PMC8295288 DOI: 10.1038/s41467-021-24608-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/17/2021] [Indexed: 01/07/2023] Open
Abstract
Latent human cytomegalovirus (HCMV) infection is characterized by limited gene expression, making latent HCMV infections refractory to current treatments targeting viral replication. However, reactivation of latent HCMV in immunosuppressed solid organ and stem cell transplant patients often results in morbidity. Here, we report the killing of latently infected cells via a virus-specific nanobody (VUN100bv) that partially inhibits signaling of the viral receptor US28. VUN100bv reactivates immediate early gene expression in latently infected cells without inducing virus production. This allows recognition and killing of latently infected monocytes by autologous cytotoxic T lymphocytes from HCMV-seropositive individuals, which could serve as a therapy to reduce the HCMV latent reservoir of transplant patients.
Collapse
|
13
|
Feng L, Li W, Wu X, Li X, Yang X, Ran Y, Wu J, Li H. Human Cytomegalovirus UL23 Attenuates Signal Transducer and Activator of Transcription 1 Phosphorylation and Type I Interferon Response. Front Microbiol 2021; 12:692515. [PMID: 34305856 PMCID: PMC8301221 DOI: 10.3389/fmicb.2021.692515] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV), the human beta-herpesvirus, can cause severe syndromes among both immunocompromised adult patients and newborns. Type I interferon (IFN-I) exerts an important effect to resist infections caused by viruses such as HCMV, while IFN evasion may serve as a key determining factor for viral dissemination and disease occurrence within hosts. In this study, UL23, a tegument protein of HCMV, was confirmed to be a key factor for negatively regulating the type I IFN immune response. A detailed analysis indicated that the viral UL23 protein increases the IFN-I antiviral resistance during HCMV infections. Furthermore, UL23 was shown to significantly reduce the levels of IFN-stimulated genes (ISGs) and promoter activity of IFN-I-stimulated response element. Mechanically, UL23 was discovered to impair the signal transducer and activator of transcription 1 (STAT1) phosphorylation, although it was not found to affect phosphorylation and expression of STAT2, Janus activated kinase 1, or tyrosine kinase 2, which are associated with IFN-I signal transduction pathway. Additionally, a significantly reduced nuclear expression of STAT1 but not of IFN regulatory factor 9 or STAT2 was observed. Findings of this study indicate that HCMV UL23 is a viral antagonist that acts against the cellular innate immunity and reveal a possible novel effect of UL23 on IFN-I signaling.
Collapse
Affiliation(s)
- Linyuan Feng
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Wanwei Li
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xingyuan Wu
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xiaotian Li
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xiaoping Yang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yanhong Ran
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jianguo Wu
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China.,Foshan Institute of Medical Microbiology, Foshan, China
| | - Hongjian Li
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| |
Collapse
|
14
|
Verweij MC, Hansen SG, Iyer R, John N, Malouli D, Morrow D, Scholz I, Womack J, Abdulhaqq S, Gilbride RM, Hughes CM, Ventura AB, Ford JC, Selseth AN, Oswald K, Shoemaker R, Berkemeier B, Bosche WJ, Hull M, Shao J, Sacha JB, Axthelm MK, Edlefsen PT, Lifson JD, Picker LJ, Früh K. Modulation of MHC-E transport by viral decoy ligands is required for RhCMV/SIV vaccine efficacy. Science 2021; 372:eabe9233. [PMID: 33766941 PMCID: PMC8354429 DOI: 10.1126/science.abe9233] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/15/2021] [Indexed: 12/15/2022]
Abstract
Strain 68-1 rhesus cytomegalovirus (RhCMV) vectors expressing simian immunodeficiency virus (SIV) antigens elicit CD8+ T cells recognizing epitopes presented by major histocompatibility complex II (MHC-II) and MHC-E but not MHC-Ia. These immune responses mediate replication arrest of SIV in 50 to 60% of monkeys. We show that the peptide VMAPRTLLL (VL9) embedded within the RhCMV protein Rh67 promotes intracellular MHC-E transport and recognition of RhCMV-infected fibroblasts by MHC-E-restricted CD8+ T cells. Deletion or mutation of viral VL9 abrogated MHC-E-restricted CD8+ T cell priming, resulting in CD8+ T cell responses exclusively targeting MHC-II-restricted epitopes. These responses were comparable in magnitude and differentiation to responses elicited by 68-1 vectors but did not protect against SIV. Thus, Rh67-enabled direct priming of MHC-E-restricted T cells is crucial for RhCMV/SIV vaccine efficacy.
Collapse
Affiliation(s)
- Marieke C Verweij
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Ravi Iyer
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Nessy John
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - David Morrow
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Isabel Scholz
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jennie Womack
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Shaheed Abdulhaqq
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Colette M Hughes
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Abigail B Ventura
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Julia C Ford
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Andrea N Selseth
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Kelli Oswald
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - Rebecca Shoemaker
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - Brian Berkemeier
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - William J Bosche
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - Michael Hull
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - Jason Shao
- Population Sciences and Computational Biology Programs, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jonah B Sacha
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael K Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Paul T Edlefsen
- Population Sciences and Computational Biology Programs, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
15
|
Barbet G, Nair-Gupta P, Schotsaert M, Yeung ST, Moretti J, Seyffer F, Metreveli G, Gardner T, Choi A, Tortorella D, Tampé R, Khanna KM, García-Sastre A, Blander JM. TAP dysfunction in dendritic cells enables noncanonical cross-presentation for T cell priming. Nat Immunol 2021; 22:497-509. [PMID: 33790474 PMCID: PMC8981674 DOI: 10.1038/s41590-021-00903-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 02/22/2021] [Indexed: 02/01/2023]
Abstract
Classic major histocompatibility complex class I (MHC-I) presentation relies on shuttling cytosolic peptides into the endoplasmic reticulum (ER) by the transporter associated with antigen processing (TAP). Viruses disable TAP to block MHC-I presentation and evade cytotoxic CD8+ T cells. Priming CD8+ T cells against these viruses is thought to rely solely on cross-presentation by uninfected TAP-functional dendritic cells. We found that protective CD8+ T cells could be mobilized during viral infection even when TAP was absent in all hematopoietic cells. TAP blockade depleted the endosomal recycling compartment of MHC-I molecules and, as such, impaired Toll-like receptor-regulated cross-presentation. Instead, MHC-I molecules accumulated in the ER-Golgi intermediate compartment (ERGIC), sequestered away from Toll-like receptor control, and coopted ER-SNARE Sec22b-mediated vesicular traffic to intersect with internalized antigen and rescue cross-presentation. Thus, when classic MHC-I presentation and endosomal recycling compartment-dependent cross-presentation are impaired in dendritic cells, cell-autonomous noncanonical cross-presentation relying on ERGIC-derived MHC-I counters TAP dysfunction to nevertheless mediate CD8+ T cell priming.
Collapse
Affiliation(s)
- Gaëtan Barbet
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- The Child Health Institute of New Jersey, and Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Priyanka Nair-Gupta
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Janssen Research and Development LLC, Spring House, PA, USA
| | - Michael Schotsaert
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephen T Yeung
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Infectious Disease, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Julien Moretti
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Fabian Seyffer
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Giorgi Metreveli
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas Gardner
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, New York, NY, USA
- ArsenalBio, San Francisco, CA, USA
| | - Angela Choi
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Moderna Inc., Cambridge, MA, USA
| | - Domenico Tortorella
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Kamal M Khanna
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Adolfo García-Sastre
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J Magarian Blander
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
16
|
Zhang Q, Song X, Ma P, Lv L, Zhang Y, Deng J, Zhang Y. Human Cytomegalovirus miR-US33as-5p Targets IFNAR1 to Achieve Immune Evasion During Both Lytic and Latent Infection. Front Immunol 2021; 12:628364. [PMID: 33746965 PMCID: PMC7973039 DOI: 10.3389/fimmu.2021.628364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/04/2021] [Indexed: 11/21/2022] Open
Abstract
As the first line of antiviral defense, type I interferon (IFN) binds IFN receptor 1 (IFNAR1) and IFNAR2 to activate the Jak-STAT signal transduction pathway, producing IFN-stimulated genes (ISGs) to control viral infection. The mechanisms by which human cytomegalovirus (HCMV) counteracts the IFN pathway are only partially defined. We show that miR-US33as-5p encoded by HCMV is expressed in both lytic and latent infection. By analysis with RNA hybrid and screening with luciferase reporter assays, we identified IFNAR1 as a target of hcmv-miR-US33as-5p, which was further verified by examining the expression of two IFNAR1 mutants and the binding of IFNAR1 to miR-US33as-5p/miR-US33as-5p-M1/miR-US33as-5p-M2. We found that after the transfection of miR-US33as-5p mimics into different cell lines, the phosphorylation of downstream proteins and ISG expression were downregulated. Immunofluorescence showed that the miR-US33as-5p mimics also inhibited STAT1 translocation into the nucleus. Furthermore, we constructed HCMV with mutant miR-US33as-5p and determined that the mutation did not affect HCMV replication. We found that MRC-5/human foreskin fibroblast (HFF) cells infected with ΔmiRNA HCMV exhibited higher IFNAR1 and ISG expression and a reduced viral load in the presence of exogenous IFN than cells infected with WT HCMV did, confirming that the knockout of miR-US33as-5p impaired viral resistance to IFN. Finally, we tested the effect of ΔmiRNA HCMV on THP-1 and d-THP-1 cells, common in vitro models of latent infection and reactivation, respectively. Again, we found that cells infected with ΔmiRNA HCMV showed a reduced viral load in the presence of IFN than the control cells did, confirming that miR-US33as-5p also affects IFN resistance during both latency and reactivation. These results indicate a new microRNA (miRNA)-based immune evasion mechanism employed by HCMV to achieve lifelong infection.
Collapse
Affiliation(s)
- Qian Zhang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| | - Xin Song
- Department of Otolaryngology Head and Neck Surgery, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Ping Ma
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| | - Liping Lv
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| | - Yangyang Zhang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| | - Jiang Deng
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| | - Yanyu Zhang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| |
Collapse
|
17
|
Noguchi K, Majima R, Takahashi K, Iwase Y, Yamada S, Satoh K, Koshizuka T, Inoue N. Identification and functional analyses of a cell-death inhibitor encoded by guinea pig cytomegalovirus gp38.1 in cell culture and in animals. J Gen Virol 2020; 101:1270-1279. [PMID: 32915127 DOI: 10.1099/jgv.0.001493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cytomegaloviruses (CMVs) employ an array of strategies designed to interfere with host defence responses against pathogens. Studies on such evasion mechanisms are important for understanding the pathogenesis of CMV diseases. Although guinea pig CMV (GPCMV) provides a useful animal model for congenital CMV infection, its evasion strategies are not fully elucidated. Here, we analysed a genome locus that may encode gene products for the GPCMV evasion mechanisms and found the following. (1) RACE analyses identified five transcripts in the GP38-gp38.4 locus, one of which was a spliced product encoding gp38.1. Similarities in the splicing pattern and gene position of gp38.1 to human CMV UL37 and its exon 1 encoding vMIA (viral mitochondria-localized inhibitor of apoptosis) suggest that the gp38.1 gene encodes an apoptosis inhibitor. (2) In a transient transfection assay, gp38.1 localized in the mitochondria and relocated BAX from the cytoplasm to the mitochondria, although its co-localization with BAK was not evident. Further, the expression of gp38.1 partially reduced staurosporine-induced apoptosis. (3) GPCMV defective in the gp38.1 ORF (Δ38.1) and the virus that rescues the defect (r38.1) were generated. Guinea pig fibroblast cells infected with Δ38.1 died earlier than r38.1-infected cells, which resulted in the lower yields of Δ38.1. (4) In animals, viral loads in the spleens of r38.1-infected guinea pigs were higher than those in the spleens of Δ38.1-infected animals. In conclusion, although GPCMV gp38.1 exerts a vMIA-like function, its inhibitory effect was not robust, suggesting the presence of additional inhibitory molecule(s), such as a BAK-specific inhibitor.
Collapse
Affiliation(s)
- Kazuma Noguchi
- Present address: Kaken Pharmaceutical, Tokyo, Japan
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Ryuichi Majima
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Keita Takahashi
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Yoshihiko Iwase
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Souichi Yamada
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Keisuke Satoh
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Tetsuo Koshizuka
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Naoki Inoue
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
18
|
Impact of Natural Occurring ERAP1 Single Nucleotide Polymorphisms within miRNA-Binding Sites on HCMV Infection. Int J Mol Sci 2020; 21:ijms21165861. [PMID: 32824160 PMCID: PMC7461596 DOI: 10.3390/ijms21165861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a β-herpesvirus that causes serious problems in people with a compromised immune system, whereas it coexists asymptomatically within the host with a healthy immune system. Like other viruses, HCMV has adopted multiples strategies to manipulate the host’s immune responses. Among them, expression of viral microRNAs (miRNAs) is one of the most intriguing. HCMV miR-UL112-5p and miR-US4-1 have been found to contribute to immune evasion by targeting the endoplasmic reticulum aminopeptidase 1 (ERAP1), a highly polymorphic key component of antigen processing. The current incomplete picture on the interplay between viral miRNAs and host immunity implies the need to better characterize the host genetic determinants. Naturally occurring single nucleotide polymorphisms (SNPs) within the miRNA binding sites of target genes may affect miRNA–target interactions. In this review, we focus on the relevance of 3′ untranslated region (3′UTR) ERAP1 SNPs within miRNA binding sites in modulating miRNA–mRNA interactions and the possible consequent individual susceptibility to HCMV infection. Moreover, we performed an in silico analysis using different bioinformatic algorithms to predict ERAP1 variants with a putative powerful biological function. This evidence provides a basis to deepen the knowledge on how 3′UTR ERAP1 variants may alter the mechanism of action of HCMV miRNAs, in order to develop targeted antiviral therapies.
Collapse
|
19
|
Xiao J, Deng J, Zhang Q, Ma P, Lv L, Zhang Y, Li C, Zhang Y. Targeting human cytomegalovirus IE genes by CRISPR/Cas9 nuclease effectively inhibits viral replication and reactivation. Arch Virol 2020; 165:1827-1835. [PMID: 32507978 DOI: 10.1007/s00705-020-04687-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/29/2020] [Indexed: 01/23/2023]
Abstract
Human cytomegalovirus (HCMV) infection causes high morbidity and mortality among immunocompromised patients and can remain in a latent state in host cells. Expression of the immediate-early (IE) genes sustains HCMV replication and reactivation. As a novel genome-editing tool, the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) system has been extensively utilized to modify and edit genomic DNA. In the present study, the CRISPR/Cas9 system was used to target the IE region of the HCMV genome via specific single-guide RNAs (sgRNAs). Infection with CRISPR/Cas9/sgRNA lentiviral constructs significantly reduced viral gene expression and virion production in HFF primary fibroblasts and inhibited viral DNA production and reactivation in the THP-1 monocytic cell line. Thus, the CRISPR/Cas9/sgRNA system can accurately and efficiently target HCMV replication and reactivation and represents a novel therapeutic strategy against latent HCMV infection.
Collapse
Affiliation(s)
- Jun Xiao
- Department of Blood Transfusion, Air Force Medical Center, PLA, 30 Fucheng Road, Beijing, 100142, P.R. China
| | - Jiang Deng
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P.R. China
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P.R. China
| | - Qian Zhang
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P.R. China
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P.R. China
| | - Ping Ma
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P.R. China
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P.R. China
| | - Liping Lv
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P.R. China
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P.R. China
| | - Yangyang Zhang
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P.R. China
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P.R. China
| | - Cuiying Li
- Department of Blood Transfusion, Air Force Medical Center, PLA, 30 Fucheng Road, Beijing, 100142, P.R. China.
| | - Yanyu Zhang
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P.R. China.
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P.R. China.
| |
Collapse
|
20
|
Poole E, Neves TC, Oliveira MT, Sinclair J, da Silva MCC. Human Cytomegalovirus Interleukin 10 Homologs: Facing the Immune System. Front Cell Infect Microbiol 2020; 10:245. [PMID: 32582563 PMCID: PMC7296156 DOI: 10.3389/fcimb.2020.00245] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Human Cytomegalovirus (HCMV) can cause a variety of health disorders that can lead to death in immunocompromised individuals and neonates. The HCMV lifecycle comprises both a lytic (productive) and a latent (non-productive) phase. HCMV lytic infection occurs in a wide range of terminally differentiated cell types. HCMV latency has been less well-studied, but one characterized site of latency is in precursor cells of the myeloid lineage. All known viral genes are expressed during a lytic infection and a subset of these are also transcribed during latency. The UL111A gene which encodes the viral IL-10, a homolog of the human IL-10, is one of these genes. During infection, different transcript isoforms of UL111A are generated by alternative splicing. The most studied of the UL111A isoforms are cmvIL-10 (also termed the "A" transcript) and LAcmvIL-10 (also termed the "B" transcript), the latter being a well-characterized latency associated transcript. Both isoforms can downregulate MHC class II, however they differ in a number of other immunomodulatory properties, such as the ability to bind the IL10 receptor and induce signaling through STAT3. There are also a number of other isoforms which have been identified which are expressed by differential splicing during lytic infection termed C, D, E, F, and G, although these have been less extensively studied. HCMV uses the viral IL-10 proteins to manipulate the immune system during lytic and latent phases of infection. In this review, we will discuss the literature on the viral IL-10 transcripts identified to date, their encoded proteins and the structures of these proteins as well as the functional properties of all the different isoforms of viral IL-10.
Collapse
Affiliation(s)
- Emma Poole
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tainan Cerqueira Neves
- Center for Natural and Humanities Sciences, Federal University of ABC (UFABC), São Bernardo do Campo, Brazil
| | - Martha Trindade Oliveira
- Center for Natural and Humanities Sciences, Federal University of ABC (UFABC), São Bernardo do Campo, Brazil
| | - John Sinclair
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
21
|
Shan L, Li S, Meeldijk J, Blijenberg B, Hendriks A, van Boxtel KJWM, van den Berg SPH, Groves IJ, Potts M, Svrlanska A, Stamminger T, Wills MR, Bovenschen N. Killer cell proteases can target viral immediate-early proteins to control human cytomegalovirus infection in a noncytotoxic manner. PLoS Pathog 2020; 16:e1008426. [PMID: 32282833 PMCID: PMC7179929 DOI: 10.1371/journal.ppat.1008426] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 04/23/2020] [Accepted: 02/21/2020] [Indexed: 12/17/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the most frequent viral cause of congenital defects and can trigger devastating disease in immune-suppressed patients. Cytotoxic lymphocytes (CD8+ T cells and NK cells) control HCMV infection by releasing interferon-γ and five granzymes (GrA, GrB, GrH, GrK, GrM), which are believed to kill infected host cells through cleavage of intracellular death substrates. However, it has recently been demonstrated that the in vivo killing capacity of cytotoxic T cells is limited and multiple T cell hits are required to kill a single virus-infected cell. This raises the question whether cytotoxic lymphocytes can use granzymes to control HCMV infection in a noncytotoxic manner. Here, we demonstrate that (primary) cytotoxic lymphocytes can block HCMV dissemination independent of host cell death, and interferon-α/β/γ. Prior to killing, cytotoxic lymphocytes induce the degradation of viral immediate-early (IE) proteins IE1 and IE2 in HCMV-infected cells. Intriguingly, both IE1 and/or IE2 are directly proteolyzed by all human granzymes, with GrB and GrM being most efficient. GrB and GrM cleave IE1 after Asp398 and Leu414, respectively, likely resulting in IE1 aberrant cellular localization, IE1 instability, and functional impairment of IE1 to interfere with the JAK-STAT signaling pathway. Furthermore, GrB and GrM cleave IE2 after Asp184 and Leu173, respectively, resulting in IE2 aberrant cellular localization and functional abolishment of IE2 to transactivate the HCMV UL112 early promoter. Taken together, our data indicate that cytotoxic lymphocytes can also employ noncytotoxic ways to control HCMV infection, which may be explained by granzyme-mediated targeting of indispensable viral proteins during lytic infection. Human cytomegalovirus (HCMV) is the leading viral cause of congenital defects, can trigger disease in immune-compromised patients, and plays roles in cancer development. Cytotoxic lymphocytes kill HCMV-infected cells via releasing a set of five cytotoxic serine proteases called granzymes. However, the killing capacity of cytotoxic cells is limited and multiple T cell hits are required to kill a single virus-infected cell. This raises the question whether cytotoxic lymphocytes can use granzymes to control HCMV infection in a noncytotoxic manner. Here, we show that cytotoxic lymphocytes can also use granzymes to inhibit HCMV replication in absence of cell death. All five granzymes cleave and inactivate both viral immediate-early (IE1/2) proteins, which are essential players for initiating HCMV infection. Our data support the model that cytotoxic cells employ granzymes to dampen HCMV replication prior to accumulation of sufficient hits to kill the infected cell.
Collapse
Affiliation(s)
- Liling Shan
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Shuang Li
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan Meeldijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bernadet Blijenberg
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Astrid Hendriks
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Ian J. Groves
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Martin Potts
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Adriana Svrlanska
- Institute of Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | - Mark R. Wills
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
22
|
Fonseca S, Pereira V, Lau C, Teixeira MDA, Bini-Antunes M, Lima M. Human Peripheral Blood Gamma Delta T Cells: Report on a Series of Healthy Caucasian Portuguese Adults and Comprehensive Review of the Literature. Cells 2020; 9:cells9030729. [PMID: 32188103 PMCID: PMC7140678 DOI: 10.3390/cells9030729] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/06/2020] [Accepted: 03/13/2020] [Indexed: 12/20/2022] Open
Abstract
Gamma delta T cells (Tc) are divided according to the type of Vδ and Vγ chains they express, with two major γδ Tc subsets being recognized in humans: Vδ2Vγ9 and Vδ1. Despite many studies in pathological conditions, only a few have quantified the γδ Tc subsets in healthy adults, and a comprehensive review of the factors influencing its representation in the blood is missing. Here we quantified the total γδ Tc and the Vδ2/Vγ9 and Vδ1 Tc subsets in the blood from 30 healthy, Caucasian, Portuguese adults, we characterized their immunophenotype by 8-color flow cytometry, focusing in a few relevant Tc markers (CD3/TCR-γδ, CD5, CD8), and costimulatory (CD28), cytotoxic (CD16) and adhesion (CD56) molecules, and we examined the impacts of age and gender. Additionally, we reviewed the literature on the influences of race/ethnicity, age, gender, special periods of life, past infections, diet, medications and concomitant diseases on γδ Tc and their subsets. Given the multitude of factors influencing the γδ Tc repertoire and immunophenotype and the high variation observed, caution should be taken in interpreting “abnormal” γδ Tc values and repertoire deviations, and the clinical significance of small populations of “phenotypically abnormal” γδ Tc in the blood.
Collapse
Affiliation(s)
- Sónia Fonseca
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP); 4099-001 Porto Porto, Portugal; (S.F.); (C.L.); (M.d.A.T.)
| | - Vanessa Pereira
- Department of Clinical Pathology, Centro Hospitalar de Vila Nova de Gaia/Espinho (CHVNG/E); 4434-502 Vila Nova de Gaia, Portugal;
| | - Catarina Lau
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP); 4099-001 Porto Porto, Portugal; (S.F.); (C.L.); (M.d.A.T.)
| | - Maria dos Anjos Teixeira
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP); 4099-001 Porto Porto, Portugal; (S.F.); (C.L.); (M.d.A.T.)
| | - Marika Bini-Antunes
- Laboratory of Immunohematology and Blood Donors Unit, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP); 4099-001Porto, Portugal;
| | - Margarida Lima
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP); 4099-001 Porto Porto, Portugal; (S.F.); (C.L.); (M.d.A.T.)
- Correspondence: ; Tel.: + 351-22-20-77-500
| |
Collapse
|
23
|
Kalejta RF, Albright ER. Expanding the Known Functional Repertoire of the Human Cytomegalovirus pp71 Protein. Front Cell Infect Microbiol 2020; 10:95. [PMID: 32226778 PMCID: PMC7080695 DOI: 10.3389/fcimb.2020.00095] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 02/25/2020] [Indexed: 12/11/2022] Open
Abstract
The human cytomegalovirus pp71 protein is packaged within the tegument of infectious virions and performs multiple functions in host cells to prime them for productive, lytic replication. Here we review the known and hypothesized functions of pp71 in regulating proteolysis, infection outcome (lytic or latent), histone deposition, transcription, translation, immune evasion, cell cycle progression, and pathogenesis. We also highlight recent advances in CMV-based vaccine candidates informed by an improved understanding of pp71 function.
Collapse
Affiliation(s)
| | - Emily R. Albright
- McArdle Laboratory for Cancer Research, Institute for Molecular Virology, University of Wisconsin – Madison, Madison, WI, United States
| |
Collapse
|
24
|
Zimmermann C, Krämer N, Krauter S, Strand D, Sehn E, Wolfrum U, Freiwald A, Butter F, Plachter B. Autophagy interferes with human cytomegalovirus genome replication, morphogenesis, and progeny release. Autophagy 2020; 17:779-795. [PMID: 32079454 DOI: 10.1080/15548627.2020.1732686] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Viral infections are often accompanied by the induction of autophagy as an intrinsic cellular defense mechanism. Herpesviruses have developed strategies to evade autophagic degradation and to manipulate autophagy of the host cells to their benefit. Here we addressed the role of macroautophagy/autophagy in human cytomegalovirus replication and for particle morphogenesis. We found that proteins of the autophagy machinery localize to cytoplasmic viral assembly compartments and enveloped virions in the cytoplasm. Surprisingly, the autophagy receptor SQSTM1/p62 was also found to colocalize with HCMV capsids in the nucleus of infected cells. This finding indicates that the autophagy machinery interacts with HCMV already at the early nuclear stages of particle morphogenesis. The membrane-bound form of LC3 and several autophagy receptors were packaged into extracellular HCMV virions. This suggested that autophagic membranes were included during secondary envelopment of HCMV virions. To further address the importance of autophagy in HCMV infection, we generated an HCMV mutant that expressed a dominant-negative version of the protease ATG4B (BAD-ATG4BC74A). The proteolytic activity of ATG4B is required for LC3 cleavage, priming it for membrane conjugation. Surprisingly, both genome replication and virus release were enhanced in cells infected with BAD-ATG4BC74A, compared to control strains. These results show that autophagy operates as an antiviral process during HCMV infection but is dispensable for secondary HCMV particle envelopment.Abbreviations: ATG: autophagy-related; BAC: bacterial artificial chromosome; BECN1: beclin 1; CPE: cytopathic effect; cVACs: cytoplasmic viral assembly compartments; d.p.i.: days post-infection; DB: dense body; EBV: Epstein-Barr virus; galK: galactokinase; HCMV: human cytomegalovirus; HFF: human foreskin fibroblasts; IE: immediate-early; IRS: internal repeat short; LC3: MAP1LC3A/B; m.o.i.; multiplicity of infection; MCP: major capsid protein; Pp: phosphoprotein; sCP/UL48a: smallest capsid protein; TRS: terminal repeat short; UL: unique long; US: unique short.
Collapse
Affiliation(s)
- Christine Zimmermann
- Institute for Virology , University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Nadine Krämer
- Institute for Virology , University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Steffi Krauter
- Institute for Virology , University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Dennis Strand
- I. Medical Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Elisabeth Sehn
- Institute of Molecular Physiology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Anja Freiwald
- Institute for Molecular Biology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Falk Butter
- Institute for Molecular Biology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Bodo Plachter
- Institute for Virology , University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
25
|
Abstract
The etiopathogenesis of severe periodontitis includes herpesvirus-bacteria coinfection. This article evaluates the pathogenicity of herpesviruses (cytomegalovirus and Epstein-Barr virus) and periodontopathic bacteria (Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis) and coinfection of these infectious agents in the initiation and progression of periodontitis. Cytomegalovirus and A. actinomycetemcomitans/P. gingivalis exercise synergistic pathogenicity in the development of localized ("aggressive") juvenile periodontitis. Cytomegalovirus and Epstein-Barr virus are associated with P. gingivalis in adult types of periodontitis. Periodontal herpesviruses that enter the general circulation may also contribute to disease development in various organ systems. A 2-way interaction is likely to occur between periodontal herpesviruses and periodontopathic bacteria, with herpesviruses promoting bacterial upgrowth, and bacterial factors reactivating latent herpesviruses. Bacterial-induced gingivitis may facilitate herpesvirus colonization of the periodontium, and herpesvirus infections may impede the antibacterial host defense and alter periodontal cells to predispose for bacterial adherence and invasion. Herpesvirus-bacteria synergistic interactions, are likely to comprise an important pathogenic determinant of aggressive periodontitis. However, mechanistic investigations into the molecular and cellular interaction between periodontal herpesviruses and bacteria are still scarce. Herpesvirus-bacteria coinfection studies may yield significant new discoveries of pathogenic determinants, and drug and vaccine targets to minimize or prevent periodontitis and periodontitis-related systemic diseases.
Collapse
Affiliation(s)
- Casey Chen
- Division of Periodontology, Diagnostic Sciences & Dental Hygiene, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Jørgen Slots
- Division of Periodontology, Diagnostic Sciences & Dental Hygiene, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
26
|
Abstract
The human betaherpesviruses, human cytomegalovirus (HCMV; species Human betaherpesvirus 5) and human herpesviruses 6A, 6B, and 7 (HHV-6A, -6B, and -7; species Human betaherpesviruses 6A, 6B, and 7) are highly prevalent and can cause severe disease in immune-compromised and immune-naive populations in well- and under-developed communities. Herpesvirus virion assembly is an intricate process that requires viral orchestration of host systems. In this review, we describe recent advances in some of the many cellular events relevant to assembly and egress of betaherpesvirus virions. These include modifications of host metabolic, immune, and autophagic/recycling systems. In addition, we discuss unique aspects of betaherpesvirus virion structure, virion assembly, and the cellular pathways employed during virion egress.
Collapse
|
27
|
Feng XG, Lin GY, Tang YS, Wu PF. Cytomegalovirus-associated acute respiratory failure in lupus patients: case reports in relation to cytomegalovirus biology. Lupus 2019; 28:1354-1359. [PMID: 31551032 DOI: 10.1177/0961203319876990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
With the wide use of immunosuppressive agents, life-threatening cytomegalovirus-associated acute respiratory failure occurs frequently. However, this condition is yet to be fully recognized and the therapeutic approach to it can only be based on comprehensive protocols rather than the biological characteristics of cytomegalovirus. We describe three acute respiratory failure events that were related to the pathogenicity of cytomegalovirus, the primary cytopathic effect and secondary antiviral immunity-mediated damage. All cytomegalovirus infection occurred after immunosuppressive usage while the acute respiratory failure events took place in different clinical settings. The first acute respiratory failure event originated from the immunoinflammatory response after cytomegalovirus infection was circumscribed, while the second resulted from cytomegalovirus reactivation and the third was caused by the combined effect of acute cytomegalovirus infection and the subsequent immunoinflammatory response. According to the clinical setting, corresponding therapeutic approaches (sequential or combined strategy) were carried out. All the patients here presented were responsive to the above therapeutic strategies. Consequently, cytomegalovirus-associated acute respiratory failure in systemic lupus erythematosus patients should be carefully differentiated and a sequential or combined strategy should be carried out according to the clinical setting. Overall, we find that there are three patterns of cytomegalovirus-associated acute respiratory failure in systemic lupus erythematosus patients and propose a novel therapeutic strategy in relation to cytomegalovirus biology.
Collapse
Affiliation(s)
- X G Feng
- Department of Rheumatology, 900th Hospital, Fujian Medical University, Fuzhou, China
| | - G Y Lin
- Department of Rheumatology, 900th Hospital, Fujian Medical University, Fuzhou, China
| | - Y S Tang
- Department of Rheumatology, 900th Hospital, Fujian Medical University, Fuzhou, China
| | - P F Wu
- Department of Rheumatology, 900th Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
28
|
Cytomegalovirus is a tumor-associated virus: armed and dangerous. Curr Opin Virol 2019; 39:49-59. [PMID: 31525538 DOI: 10.1016/j.coviro.2019.08.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/05/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022]
Abstract
Human cytomegalovirus (HCMV) gene products are present in multiple human malignancies, often in specific association with tumor cells and tumor vasculature. Emerging evidence from human and mouse models of CMV infection in cancer indicate that CMV can transform epithelial cells, promote epithelial to mesenchymal transition (EMT) and mesenchymal to epithelial (MET) in tumor cells, promote tumor angiogenesis and proliferation and incapacitate the host anti-CMV immune response. This review will discuss the increasing role of HCMV in human cancer by demonstrating how HCMV is well suited for impacting major themes in oncogenesis including initiation, promotion, progression, metastasis and immune evasion. What emerges is a picture of an extremely versatile pathogen that may play a significant role in human cancer progression and death.
Collapse
|
29
|
Production Strategies for Pentamer-Positive Subviral Dense Bodies as a Safe Human Cytomegalovirus Vaccine. Vaccines (Basel) 2019; 7:vaccines7030104. [PMID: 31480520 PMCID: PMC6789746 DOI: 10.3390/vaccines7030104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022] Open
Abstract
Infections with the human cytomegalovirus (HCMV) are associated with severe clinical manifestations in children following prenatal transmission and after viral reactivation in immunosuppressed individuals. The development of an HCMV vaccine has long been requested but there is still no licensed product available. Subviral dense bodies (DB) are immunogenic in pre-clinical models and are thus a promising HCMV vaccine candidate. Recently, we established a virus based on the laboratory strain Towne that synthesizes large numbers of DB containing the pentameric protein complex gH/gL/UL128-131 (Towne-UL130repΔGFP). The work presented here focuses on providing strategies for the production of a safe vaccine based on that strain. A GMP-compliant protocol for DB production was established. Furthermore, the DB producer strain Towne-UL130rep was attenuated by deleting the UL25 open reading frame. Additional genetic modifications aim to abrogate its capacity to replicate in vivo by conditionally expressing pUL51 using the Shield-1/FKBP destabilization system. We further show that the terminase inhibitor letermovir can be used to reduce infectious virus contamination of a DB vaccine by more than two orders of magnitude. Taken together, strategies are provided here that allow for the production of a safe and immunogenic DB vaccine for clinical testing.
Collapse
|
30
|
Becerra-Artiles A, Cruz J, Leszyk JD, Sidney J, Sette A, Shaffer SA, Stern LJ. Naturally processed HLA-DR3-restricted HHV-6B peptides are recognized broadly with polyfunctional and cytotoxic CD4 T-cell responses. Eur J Immunol 2019; 49:1167-1185. [PMID: 31020640 DOI: 10.1002/eji.201948126] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/13/2019] [Accepted: 04/23/2019] [Indexed: 01/06/2023]
Abstract
Human herpes virus 6B (HHV-6B) is a widespread virus that infects most people early in infancy and establishes a chronic life-long infection with periodic reactivation. CD4 T cells have been implicated in control of HHV-6B, but antigenic targets and functional characteristics of the CD4 T-cell response are poorly understood. We identified 25 naturally processed MHC-II peptides, derived from six different HHV-6B proteins, and showed that they were recognized by CD4 T-cell responses in HLA-matched donors. The peptides were identified by mass spectrometry after elution from HLA-DR molecules isolated from HHV-6B-infected T cells. The peptides showed strong binding to matched HLA alleles and elicited recall T-cell responses in vitro. T-cell lines expanded in vitro were used for functional characterization of the response. Responding cells were mainly CD3+ CD4+ , produced IFN-γ, TNF-α, and low levels of IL-2, alone or in combination, highlighting the presence of polyfunctional T cells in the overall response. Many of the responding cells mobilized CD107a, stored granzyme B, and mediated specific killing of peptide-pulsed target cells. These results highlight a potential role for polyfunctional cytotoxic CD4 T cells in the long-term control of HHV-6B infection.
Collapse
Affiliation(s)
| | - John Cruz
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA
| | - John D Leszyk
- Mass Spectrometry Facility, University of Massachusetts Medical School, Shrewsbury, MA.,Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Immunology, San Diego, CA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, San Diego, CA.,Department of Medicine, University of California, San Diego, CA
| | - Scott A Shaffer
- Mass Spectrometry Facility, University of Massachusetts Medical School, Shrewsbury, MA.,Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| | - Lawrence J Stern
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA.,Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
31
|
Abstract
Human cytomegalovirus (HCMV) latency and reactivation is regulated by the chromatin structure at the major immediate early promoter (MIEP) within myeloid cells. Both cellular and viral factors are known to control this promoter during latency, here we will review the known mechanisms for MIEP regulation during latency. We will then focus on the virally encoded G-protein coupled receptor, US28, which suppresses the MIEP in early myeloid lineage cells. The importance of this function is underlined by the fact that US28 is essential for HCMV latency in CD34+ progenitor cells and CD14+ monocytes. We will describe cellular signalling pathways modulated by US28 to direct MIEP suppression during latency and demonstrate how US28 is able to ‘regulate the regulators’ of HCMV latency. Finally, we will describe how cell-surface US28 can be a target for antiviral therapies directed at the latent viral reservoir.
Collapse
|
32
|
Botto S, Abraham J, Mizuno N, Pryke K, Gall B, Landais I, Streblow DN, Fruh KJ, DeFilippis VR. Human Cytomegalovirus Immediate Early 86-kDa Protein Blocks Transcription and Induces Degradation of the Immature Interleukin-1β Protein during Virion-Mediated Activation of the AIM2 Inflammasome. mBio 2019; 10:e02510-18. [PMID: 30755509 PMCID: PMC6372796 DOI: 10.1128/mbio.02510-18] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022] Open
Abstract
Secretion of interleukin-1β (IL-1β) represents a fundamental innate immune response to microbial infection that, at the molecular level, occurs following activation of proteolytic caspases that cleave the immature protein into a secretable form. Human cytomegalovirus (HCMV) is the archetypal betaherpesvirus that is invariably capable of lifelong infection through the activity of numerous virally encoded immune evasion phenotypes. Innate immune pathways responsive to cytoplasmic double-stranded DNA (dsDNA) are known to be activated in response to contact between HCMV and host cells. Here, we used clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR-associated protein 9 (Cas9) genome editing to demonstrate that the dsDNA receptor absent in melanoma 2 (AIM2) is required for secretion of IL-1β following HCMV infection. Furthermore, dsDNA-responsive innate signaling induced by HCMV infection that leads to activation of the type I interferon response is also shown, unexpectedly, to play a contributory role in IL-1β secretion. Importantly, we also show that rendering virus particles inactive by UV exposure leads to substantially increased IL-1β processing and secretion and that live HCMV can inhibit this, suggesting the virus encodes factors that confer an inhibitory effect on this response. Further examination revealed that ectopic expression of the immediate early (IE) 86-kDa protein (IE86) is actually associated with a block in transcription of the pro-IL-1β gene and, independently, diminishment of the immature protein. Overall, these results reveal two new and distinct phenotypes conferred by the HCMV IE86 protein, as well as an unusual circumstance in which a single herpesviral protein exhibits inhibitory effects on multiple molecular processes within the same innate immune response.IMPORTANCE Persistent infection with HCMV is associated with the operation of diverse evasion phenotypes directed at antiviral immunity. Obstruction of intrinsic and innate immune responses is typically conferred by viral proteins either associated with the viral particle or expressed immediately after entry. In line with this, numerous phenotypes are attributed to the HCMV IE86 protein that involve interference with innate immune processes via transcriptional and protein-directed mechanisms. We describe novel IE86-mediated phenotypes aimed at virus-induced secretion of IL-1β. Intriguingly, while many viruses target the function of the molecular scaffold required for IL-1β maturation to prevent this response, we find that HCMV and IE86 target the IL-1β protein specifically. Moreover, we show that IE86 impairs both the synthesis of the IL-1β transcript and the stability of the immature protein. This indicates an unusual phenomenon in which a single viral protein exhibits two molecularly separate evasion phenotypes directed at a single innate cytokine.
Collapse
Affiliation(s)
- Sara Botto
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Jinu Abraham
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Nobuyo Mizuno
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Kara Pryke
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Bryan Gall
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Igor Landais
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Klaus J Fruh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Victor R DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
33
|
Bastidas-Legarda LY, Khakoo SI. Conserved and variable natural killer cell receptors: diverse approaches to viral infections. Immunology 2019; 156:319-328. [PMID: 30570753 DOI: 10.1111/imm.13039] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023] Open
Abstract
Natural killer (NK) cells are lymphocytes of the innate immune system with essential roles during viral infections. NK cell functions are mediated through a repertoire of non-rearranging inhibitory and activating receptors that interact with major histocompatibility complex (MHC)-peptide complexes on the surface of infected cells. Recent work studying the conserved CD94-NKG2A and variable killer cell immunoglobulin-like receptor-MHC systems suggest that these two receptor families may have subtly different properties in terms of interactions with MHC class I bound peptides, and in recognition of down-regulation of MHC class I. In this review, we discuss how these properties generate diversity in the NK cell response to viruses.
Collapse
Affiliation(s)
- Leidy Y Bastidas-Legarda
- Faculty of Medicine, Clinical and Experimental Sciences, Southampton General Hospital, University of Southampton, Southampton, UK
| | - Salim I Khakoo
- Faculty of Medicine, Clinical and Experimental Sciences, Southampton General Hospital, University of Southampton, Southampton, UK
| |
Collapse
|
34
|
Huth A, Liang X, Krebs S, Blum H, Moosmann A. Antigen-Specific TCR Signatures of Cytomegalovirus Infection. THE JOURNAL OF IMMUNOLOGY 2018; 202:979-990. [PMID: 30587531 DOI: 10.4049/jimmunol.1801401] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/19/2018] [Indexed: 11/19/2022]
Abstract
CMV is a prevalent human pathogen. The virus cannot be eliminated from the body, but is kept in check by CMV-specific T cells. Patients with an insufficient T cell response, such as transplant recipients, are at high risk of developing CMV disease. However, the CMV-specific T cell repertoire is complex, and it is not yet clear which T cells protect best against virus reactivation and disease. In this study, we present a highly resolved characterization of CMV-specific human CD8+ T cells based on enrichment by specific peptide stimulation and mRNA sequencing of their TCR β-chains (TCRβ). Our analysis included recently identified T cell epitopes restricted through HLA-C, whose presentation is resistant to viral immunomodulation, and well-studied HLA-B-restricted epitopes. In eight healthy virus carriers, we identified a total of 1052 CMV-specific TCRβ sequences. HLA-C-restricted, CMV-specific TCRβ clonotypes dominated the ex vivo T cell response and contributed the highest-frequency clonotype of the entire repertoire in two of eight donors. We analyzed sharing and similarity of CMV-specific TCRβ sequences and identified 63 public or related sequences belonging to 17 public TCRβ families. In our cohort, and in an independent cohort of 352 donors, the cumulative frequency of these public TCRβ family members was a highly discriminatory indicator of carrying both CMV infection and the relevant HLA type. Based on these findings, we propose CMV-specific TCRβ signatures as a biomarker for an antiviral T cell response to identify patients in need of treatment and to guide future development of immunotherapy.
Collapse
Affiliation(s)
- Alina Huth
- German Center for Infection Research Group Host Control of Viral Latency and Reactivation, Research Unit Gene Vectors, Helmholtz Center Munich, 81377 Munich, Germany.,Deutsches Zentrum für Infektionsforschung, 81377 Munich, Germany; and
| | - Xiaoling Liang
- German Center for Infection Research Group Host Control of Viral Latency and Reactivation, Research Unit Gene Vectors, Helmholtz Center Munich, 81377 Munich, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilian University of Munich, 81377 Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilian University of Munich, 81377 Munich, Germany
| | - Andreas Moosmann
- German Center for Infection Research Group Host Control of Viral Latency and Reactivation, Research Unit Gene Vectors, Helmholtz Center Munich, 81377 Munich, Germany; .,Deutsches Zentrum für Infektionsforschung, 81377 Munich, Germany; and
| |
Collapse
|
35
|
Wang X, Piersma SJ, Nelson CA, Dai YN, Christensen T, Lazear E, Yang L, Sluijter M, van Hall T, Hansen TH, Yokoyama WM, Fremont DH. A herpesvirus encoded Qa-1 mimic inhibits natural killer cell cytotoxicity through CD94/NKG2A receptor engagement. eLife 2018; 7:38667. [PMID: 30575523 PMCID: PMC6320069 DOI: 10.7554/elife.38667] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 12/20/2018] [Indexed: 11/13/2022] Open
Abstract
A recurrent theme in viral immune evasion is the sabotage of MHC-I antigen presentation, which brings virus the concomitant issue of ‘missing-self’ recognition by NK cells that use inhibitory receptors to detect surface MHC-I proteins. Here, we report that rodent herpesvirus Peru (RHVP) encodes a Qa-1 like protein (pQa-1) via RNA splicing to counteract NK activation. While pQa-1 surface expression is stabilized by the same canonical peptides presented by murine Qa-1, pQa-1 is GPI-anchored and resistant to the activity of RHVP pK3, a ubiquitin ligase that targets MHC-I for degradation. pQa-1 tetramer staining indicates that it recognizes CD94/NKG2A receptors. Consistently, pQa-1 selectively inhibits NKG2A+ NK cells and expression of pQa-1 can protect tumor cells from NK control in vivo. Collectively, these findings reveal an innovative NK evasion strategy wherein RHVP encodes a modified Qa-1 mimic refractory to MHC-I sabotage and capable of specifically engaging inhibitory receptors to circumvent NK activation.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Sytse J Piersma
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, United States
| | - Christopher A Nelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Ya-Nan Dai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Ted Christensen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Eric Lazear
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Liping Yang
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, United States
| | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Ted H Hansen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Wayne M Yokoyama
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States.,Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, United States
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States.,Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, United States.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, United States
| |
Collapse
|
36
|
Manickam C, Shah SV, Lucar O, Ram DR, Reeves RK. Cytokine-Mediated Tissue Injury in Non-human Primate Models of Viral Infections. Front Immunol 2018; 9:2862. [PMID: 30568659 PMCID: PMC6290327 DOI: 10.3389/fimmu.2018.02862] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022] Open
Abstract
Viral infections trigger robust secretion of interferons and other antiviral cytokines by infected and bystander cells, which in turn can tune the immune response and may lead to viral clearance or immune suppression. However, aberrant or unrestricted cytokine responses can damage host tissues, leading to organ dysfunction, and even death. To understand the cytokine milieu and immune responses in infected host tissues, non-human primate (NHP) models have emerged as important tools. NHP have been used for decades to study human infections and have played significant roles in the development of vaccines, drug therapies and other immune treatment modalities, aided by an ability to control disease parameters, and unrestricted tissue access. In addition to the genetic and physiological similarities with humans, NHP have conserved immunologic properties with over 90% amino acid similarity for most cytokines. For example, human-like symptomology and acute respiratory syndrome is found in cynomolgus macaques infected with highly pathogenic avian influenza virus, antibody enhanced dengue disease is common in neotropical primates, and in NHP models of viral hepatitis cytokine-induced inflammation induces severe liver damage, fibrosis, and hepatocellular carcinoma recapitulates human disease. To regulate inflammation, anti-cytokine therapy studies in NHP are underway and will provide important insights for future human interventions. This review will provide a comprehensive outline of the cytokine-mediated exacerbation of disease and tissue damage in NHP models of viral infections and therapeutic strategies that can aid in prevention/treatment of the disease syndromes.
Collapse
Affiliation(s)
- Cordelia Manickam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Spandan V. Shah
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Olivier Lucar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Daniel R. Ram
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - R. Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
37
|
Valencia S, Gill RB, Dowdell KC, Wang Y, Hornung R, Bowman JJ, Lacayo JC, Cohen JI. Comparison of vaccination with rhesus CMV (RhCMV) soluble gB with a RhCMV replication-defective virus deleted for MHC class I immune evasion genes in a RhCMV challenge model. Vaccine 2018; 37:333-342. [PMID: 30522906 DOI: 10.1016/j.vaccine.2018.08.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 07/06/2018] [Accepted: 08/17/2018] [Indexed: 11/25/2022]
Abstract
A human cytomegalovirus (HCMV) vaccine to prevent infection and/or reduce disease associated with congenital infection or visceral disease in transplant recipients is a high priority, but has remained elusive. We created a disabled infectious single cycle rhesus CMV (RhCMV) deleted for glycoprotein L (gL) and the MHC class I immune evasion genes Rh178 and Rh182-189, and restored its epithelial cell tropism by inserting the Rh128-131A genes. The resulting virus, RhCMVRΔgL/178/182-189, was used to vaccinate rhesus monkeys intramuscularly and was compared with vaccination of animals with soluble RhCMV glycoprotein B (gB) in alum/monophosphoryl lipid A or with PBS as a control. At 4 weeks after the second vaccination, an increased frequency of RhCMV-specific CD8 T cells was detected in animals vaccinated with the RhCMVRΔgL/178/182-189 vaccine compared to animals vaccinated with soluble gB. In contrast, monkeys vaccinated with soluble gB had 20-fold higher gB antibody titers than animals vaccinated with RhCMVRΔgL/178/182-189. Titers of neutralizing antibody to RhCMV infection of fibroblasts were higher in animals vaccinated with gB compared with RhCMVRΔgL/178/182-189. Following vaccination, monkeys were challenged subcutaneously with RhCMV UCD59, a low passage virus propagated in monkey kidney epithelial cells. All animals became infected after challenge; however, the frequency of RhCMV detection in the blood was reduced in monkeys vaccinated with soluble gB compared with those vaccinated with RhCMVRΔgL/178/182-189. The frequency of challenge virus shedding in the urine and saliva and the RhCMV copy number shed at these sites was not different in animals vaccinated with RhCMVRΔgL/178/182-189 or soluble gB compared with those that received PBS before challenge. Although the RhCMVRΔgL/178/182-189 vaccine was superior in inducing cellular immunity to RhCMV, it induced lower titers of neutralizing antibody and antibody to gB than the soluble gB vaccine; after challenge, animals vaccinated with soluble gB had a lower frequency of virus detection in the blood than those vaccinated with RhCMVRΔgL/178/182-189.
Collapse
Affiliation(s)
- Sarah Valencia
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rachel B Gill
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kennichi C Dowdell
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yanmei Wang
- Clinical Services Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Ron Hornung
- Clinical Services Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - J Jason Bowman
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juan C Lacayo
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey I Cohen
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
38
|
Theobald SJ, Khailaie S, Meyer-Hermann M, Volk V, Olbrich H, Danisch S, Gerasch L, Schneider A, Sinzger C, Schaudien D, Lienenklaus S, Riese P, Guzman CA, Figueiredo C, von Kaisenberg C, Spineli LM, Glaesener S, Meyer-Bahlburg A, Ganser A, Schmitt M, Mach M, Messerle M, Stripecke R. Signatures of T and B Cell Development, Functional Responses and PD-1 Upregulation After HCMV Latent Infections and Reactivations in Nod.Rag.Gamma Mice Humanized With Cord Blood CD34 + Cells. Front Immunol 2018; 9:2734. [PMID: 30524448 PMCID: PMC6262073 DOI: 10.3389/fimmu.2018.02734] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/06/2018] [Indexed: 12/27/2022] Open
Abstract
Human cytomegalovirus (HCMV) latency is typically harmless but reactivation can be largely detrimental to immune compromised hosts. We modeled latency and reactivation using a traceable HCMV laboratory strain expressing the Gaussia luciferase reporter gene (HCMV/GLuc) in order to interrogate the viral modulatory effects on the human adaptive immunity. Humanized mice with long-term (more than 17 weeks) steady human T and B cell immune reconstitutions were infected with HCMV/GLuc and 7 weeks later were further treated with granulocyte-colony stimulating factor (G-CSF) to induce viral reactivations. Whole body bio-luminescence imaging analyses clearly differentiated mice with latent viral infections vs. reactivations. Foci of vigorous viral reactivations were detectable in liver, lymph nodes and salivary glands. The number of viral genome copies in various tissues increased upon reactivations and were detectable in sorted human CD14+, CD169+, and CD34+ cells. Compared with non-infected controls, mice after infections and reactivations showed higher thymopoiesis, systemic expansion of Th, CTL, Treg, and Tfh cells and functional antiviral T cell responses. Latent infections promoted vast development of memory CD4+ T cells while reactivations triggered a shift toward effector T cells expressing PD-1. Further, reactivations prompted a marked development of B cells, maturation of IgG+ plasma cells, and HCMV-specific antibody responses. Multivariate statistical methods were employed using T and B cell immune phenotypic profiles obtained with cells from several tissues of individual mice. The data was used to identify combinations of markers that could predict an HCMV infection vs. reactivation status. In spleen, but not in lymph nodes, higher frequencies of effector CD4+ T cells expressing PD-1 were among the factors most suited to distinguish HCMV reactivations from infections. These results suggest a shift from a T cell dominated immune response during latent infections toward an exhausted T cell phenotype and active humoral immune response upon reactivations. In sum, this novel in vivo humanized model combined with advanced analyses highlights a dynamic system clearly specifying the immunological spatial signatures of HCMV latency and reactivations. These signatures can be merged as predictive biomarker clusters that can be applied in the clinical translation of new therapies for the control of HCMV reactivation.
Collapse
Affiliation(s)
- Sebastian J Theobald
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,Excellence Cluster REBIRTH, Laboratory of Regenerative Immune Therapies Applied, Hannover Medical School, Hannover, Germany.,Partner Site Hannover-Braunschweig, German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Sahamoddin Khailaie
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Biochemistry, Biotechnology and Bioinformatics, Technical University Braunschweig, Braunschweig, Germany
| | - Valery Volk
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,Excellence Cluster REBIRTH, Laboratory of Regenerative Immune Therapies Applied, Hannover Medical School, Hannover, Germany
| | - Henning Olbrich
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,Excellence Cluster REBIRTH, Laboratory of Regenerative Immune Therapies Applied, Hannover Medical School, Hannover, Germany.,Partner Site Hannover-Braunschweig, German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Simon Danisch
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,Excellence Cluster REBIRTH, Laboratory of Regenerative Immune Therapies Applied, Hannover Medical School, Hannover, Germany.,Partner Site Hannover-Braunschweig, German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Laura Gerasch
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,Excellence Cluster REBIRTH, Laboratory of Regenerative Immune Therapies Applied, Hannover Medical School, Hannover, Germany
| | - Andreas Schneider
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,Excellence Cluster REBIRTH, Laboratory of Regenerative Immune Therapies Applied, Hannover Medical School, Hannover, Germany
| | | | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Stefan Lienenklaus
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Peggy Riese
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research Braunschweig, Braunschweig, Germany
| | - Carlos A Guzman
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research Braunschweig, Braunschweig, Germany
| | | | | | - Loukia M Spineli
- Institute for Biostatistics, Hannover Medical School, Hannover, Germany
| | - Stephanie Glaesener
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | | | - Arnold Ganser
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Michael Schmitt
- Department of Hematology, Oncology and Rheumatology, GMP Core Facility, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Mach
- Institute of Virology, University Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Messerle
- Partner Site Hannover-Braunschweig, German Center for Infection Research (DZIF), Braunschweig, Germany.,Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Renata Stripecke
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,Excellence Cluster REBIRTH, Laboratory of Regenerative Immune Therapies Applied, Hannover Medical School, Hannover, Germany.,Partner Site Hannover-Braunschweig, German Center for Infection Research (DZIF), Braunschweig, Germany
| |
Collapse
|
39
|
Modulation of the innate immune response by human cytomegalovirus. INFECTION GENETICS AND EVOLUTION 2018; 64:105-114. [DOI: 10.1016/j.meegid.2018.06.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 06/19/2018] [Indexed: 12/19/2022]
|
40
|
A Prominent Role of the Human Cytomegalovirus UL8 Glycoprotein in Restraining Proinflammatory Cytokine Production by Myeloid Cells at Late Times during Infection. J Virol 2018; 92:JVI.02229-17. [PMID: 29467314 DOI: 10.1128/jvi.02229-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/14/2018] [Indexed: 01/21/2023] Open
Abstract
Human cytomegalovirus (HCMV) persistence in infected individuals relies on a plethora of mechanisms to efficiently reduce host immune responses. To that end, HCMV uses a variety of gene products, some of which have not been identified yet. Here we characterized the UL8 gene, which consists of two exons, sharing the first with the HCMV RL11 family member UL7 UL8 is a transmembrane protein with an N-terminal immunoglobulin (Ig)-like domain in common with UL7 but with an extended stalk and a distinctive cytoplasmic tail. The UL8 open reading frame gives rise to a heavily glycosylated protein predominantly expressed on the cell surface, from where it can be partially endocytosed and subsequently degraded. Infections with UL8-tagged viruses indicated that UL8 was synthesized with late-phase kinetics. By virtue of its highly conserved Ig-like domain, this viral protein interacted with a surface molecule present on activated neutrophils. Notably, when ectopically expressed in THP-1 myeloid cells, UL8 was able to significantly reduce the production of a variety of proinflammatory cytokines. Mutations in UL8 indicated that this functional effect was mediated by the cell surface expression of its Ig-like domain. To investigate the impact of the viral protein in the infection context, we engineered HCMVs lacking the UL8 gene and demonstrated that UL8 decreases the release of a large number of proinflammatory factors at late times after infection of THP-1 cells. Our data indicate that UL8 may exert an immunosuppressive role key for HCMV survival in the host.IMPORTANCE HCMV is a major pathogen that causes life-threatening diseases and disabilities in infected newborns and immunocompromised individuals. Containing one of the largest genomes among all reported human viruses, HCMV encodes an impressive repertoire of gene products. However, the functions of a large proportion of them still remain unknown, a fact that complicates the design of new therapeutic approaches to prevent or treat HCMV-associated diseases. In this report, we have conducted an extensive study of UL8, one of the previously uncharacterized HCMV open reading frames. We found that the UL8 protein is expressed at late times postinfection and utilized by HCMV to reduce the production of proinflammatory factors by infected myeloid cells. Thus, the work presented here points to a key role of UL8 as a novel HCMV immune modulator capable of restraining host antiviral defenses.
Collapse
|
41
|
Feng L, Sheng J, Vu GP, Liu Y, Foo C, Wu S, Trang P, Paliza-Carre M, Ran Y, Yang X, Sun X, Deng Z, Zhou T, Lu S, Li H, Liu F. Human cytomegalovirus UL23 inhibits transcription of interferon-γ stimulated genes and blocks antiviral interferon-γ responses by interacting with human N-myc interactor protein. PLoS Pathog 2018; 14:e1006867. [PMID: 29377960 PMCID: PMC5805366 DOI: 10.1371/journal.ppat.1006867] [Citation(s) in RCA: 30] [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: 01/10/2017] [Revised: 02/08/2018] [Accepted: 01/09/2018] [Indexed: 12/24/2022] Open
Abstract
Interferon-γ (IFN-γ) represents one of the most important innate immunity responses in a host to combat infections of many human viruses including human herpesviruses. Human N-myc interactor (Nmi) protein, which has been shown to interact with signal transducer and activator of transcription (STAT) proteins including STAT1, is important for the activation of IFN-γ induced STAT1-dependent transcription of many genes responsible for IFN-γ immune responses. However, no proteins encoded by herpesviruses have been reported to interact with Nmi and inhibit Nmi-mediated activation of IFN-γ immune responses to achieve immune evasion from IFN-γ responses. In this study, we show strong evidence that the UL23 protein of human cytomegalovirus (HCMV), a human herpesvirus, specifically interacts with Nmi. This interaction was identified through a yeast two-hybrid screen and co-immunoprecipitation in human cells. We observed that Nmi, when bound to UL23, was not associated with STAT1, suggesting that UL23 binding of Nmi disrupts the interaction of Nmi with STAT1. In cells overexpressing UL23, we observed (a) significantly reduced levels of Nmi and STAT1 in the nuclei, the sites where these proteins act to induce transcription of IFN-γ stimulated genes, and (b) decreased levels of the induction of the transcription of IFN-γ stimulated genes. UL23-deficient HCMV mutants induced higher transcription of IFN-γ stimulated genes and exhibited lower titers than parental and control revertant viruses expressing functional UL23 in IFN-γ treated cells. Thus, UL23 appears to interact directly with Nmi and inhibit nuclear translocation of Nmi and its associated protein STAT1, leading to a decrease of IFN-γ induced responses and an increase of viral resistance to IFN-γ. Our results further highlight the roles of UL23-Nmi interactions in facilitating viral immune escape from IFN-γ responses and enhancing viral resistance to IFN antiviral effects. Interferon-γ (IFN-γ) responses are vital for a host to combat infections of many human viruses including human herpesviruses. Upon treatment of IFN-γ, transcription of many genes responsible for IFN-γ immune responses is activated primarily by the signal transducer and activator of transcription (STAT) proteins such as STAT1 protein. Human N-myc interactor (Nmi) protein has been shown to interact with STAT proteins including STAT1 and activate IFN-γ induced STAT-dependent transcription. However, no proteins encoded by herpesviruses have been reported to interact with Nmi and inhibit Nmi-mediated activation of IFN-γ immune responses to achieve immune evasion from IFN-γ responses. In this study, we show strong evidence that the UL23 protein of human cytomegalovirus (HCMV), a human herpesvirus, specifically interacts with Nmi protein. UL23 appears to interact directly with Nmi and inhibit nuclear translocation of Nmi and its associated protein STAT1, leading to a decrease of IFN-γ responses and an increase of viral resistance to IFN-γ. Blocking UL23 expression led to higher transcription of IFN-γ stimulated genes and significant inhibition of viral growth in infected cells. These results suggest that interfering with Nmi function may represent an effective mechanism for a herpesvirus to block Nmi-mediated IFN-γ responses and increase viral resistance to IFN-γ. This also provides a potentially new therapeutic strategy to treat HCMV infection by modulating Nmi activity with blocking the expression of a viral protein.
Collapse
Affiliation(s)
- Linyuan Feng
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Jingxue Sheng
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Gia-Phong Vu
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Yujun Liu
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
- School of Medicine, St. George’s University, Grenada, West Indies
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Chingman Foo
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Songbin Wu
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Phong Trang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Marco Paliza-Carre
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Yanhong Ran
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Xiaoping Yang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Xu Sun
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Zemin Deng
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Tianhong Zhou
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Sangwei Lu
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Hongjian Li
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
- * E-mail: (FL); (HL)
| | - Fenyong Liu
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
- * E-mail: (FL); (HL)
| |
Collapse
|
42
|
Zou F, Lu ZT, Wang S, Wu S, Wu YY, Sun ZR. Human cytomegalovirus UL141 protein interacts with CELF5 and affects viral DNA replication. Mol Med Rep 2018; 17:4657-4664. [PMID: 29328469 DOI: 10.3892/mmr.2018.8419] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 01/05/2018] [Indexed: 11/06/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection is the primary viral cause of congenital abnormalities and mental retardation in newborns. The HCMV UL141‑encoded glycoprotein has been previously revealed to inhibit the cell‑surface expression of cluster of differentiation (CD)155, CD122, tumor necrosis factor‑related apoptosis‑inducing ligand death (TRAIL)‑receptor 1 (R1) and TRAIL‑receptor 2 (R2), thus protecting virally‑infected cells by allowing them to escape natural killer cell‑mediated cytotoxicity. The present study investigated the interaction between HCMV UL141 and human fetal brain cDNA to elucidate the possible effects of UL141 on the nervous system. The findings of the current study demonstrate that the HCMV UL141 protein directly interacts with the human protein CUGBP Elav‑like family member 5 (CELF5) via yeast two‑hybrid screening, this interaction was confirmed by glutathione S‑transferase pull‑down and co‑immunoprecipitation assays. Additionally, the present study demonstrated that the UL141 protein co‑localizes with CELF5 in the cytoplasm of 293 cells using fluorescence confocal microscopy. CELF5 overexpression in a stably‑expressing cell line significantly increased viral DNA copy number and titer in HCMV‑infected U373MG cells. However, reducing CELF5 expression via specific small interfering RNAs did not affect viral DNA copy number or titer in HCMV‑infected cells. The current findings suggest that the interaction between UL141 and CELF5 may be involved in modulating viral DNA synthesis and progeny production. Therefore, CELF5 may represent a possible mechanism for regulation of HCMV genomic DNA synthesis, which is a key step during HCMV infection leading to neurological disease.
Collapse
Affiliation(s)
- Fei Zou
- Department of BioBank, Affiliated Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zhi-Tao Lu
- Department of Pediatrics, Zhangjiagang First People's Hospital, Zhangjiagang, Jiangsu 215600, P.R. China
| | - Shuang Wang
- Department of BioBank, Affiliated Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Si Wu
- Department of BioBank, Affiliated Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Ying-Ying Wu
- Department of BioBank, Affiliated Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zheng-Rong Sun
- Department of BioBank, Affiliated Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
43
|
Korbecki J, Gutowska I, Kojder I, Jeżewski D, Goschorska M, Łukomska A, Lubkowska A, Chlubek D, Baranowska-Bosiacka I. New extracellular factors in glioblastoma multiforme development: neurotensin, growth differentiation factor-15, sphingosine-1-phosphate and cytomegalovirus infection. Oncotarget 2018; 9:7219-7270. [PMID: 29467963 PMCID: PMC5805549 DOI: 10.18632/oncotarget.24102] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 01/02/2018] [Indexed: 11/25/2022] Open
Abstract
Recent years have seen considerable progress in understanding the biochemistry of cancer. For example, more significance is now assigned to the tumor microenvironment, especially with regard to intercellular signaling in the tumor niche which depends on many factors secreted by tumor cells. In addition, great progress has been made in understanding the influence of factors such as neurotensin, growth differentiation factor-15 (GDF-15), sphingosine-1-phosphate (S1P), and infection with cytomegalovirus (CMV) on the 'hallmarks of cancer' in glioblastoma multiforme. Therefore, in the present work we describe the influence of these factors on the proliferation and apoptosis of neoplastic cells, cancer stem cells, angiogenesis, migration and invasion, and cancer immune evasion in a glioblastoma multiforme tumor. In particular, we discuss the effect of neurotensin, GDF-15, S1P (including the drug FTY720), and infection with CMV on tumor-associated macrophages (TAM), microglial cells, neutrophil and regulatory T cells (Treg), on the tumor microenvironment. In order to better understand the role of the aforementioned factors in tumoral processes, we outline the latest models of intratumoral heterogeneity in glioblastoma multiforme. Based on the most recent reports, we discuss the problems of multi-drug therapy in treating glioblastoma multiforme.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland.,Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biała, 43-309 Bielsko-Biała, Poland
| | - Izabela Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Ireneusz Kojder
- Department of Applied Neurocognitivistics, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland.,Department of Neurosurgery, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Dariusz Jeżewski
- Department of Applied Neurocognitivistics, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland.,Department of Neurosurgery, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Marta Goschorska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Agnieszka Łukomska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Anna Lubkowska
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| |
Collapse
|
44
|
Peppa D. Natural Killer Cells in Human Immunodeficiency Virus-1 Infection: Spotlight on the Impact of Human Cytomegalovirus. Front Immunol 2017; 8:1322. [PMID: 29089947 PMCID: PMC5650968 DOI: 10.3389/fimmu.2017.01322] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/29/2017] [Indexed: 12/14/2022] Open
Abstract
Human cytomegalovirus (HCMV) has been closely associated with the human race across evolutionary time. HCMV co-infection is nearly universal in human immunodeficiency virus-1 (HIV-1)-infected individuals and remains an important cofactor in HIV-1 disease progression even in the era of effective antiretroviral treatment. HCMV infection has been shown to have a broad and potent influence on the human immune system and has been linked with the discovery and characterization of adaptive natural killer (NK) cells. Distinct NK-cell subsets, predominately expressing the activating receptor NKG2C and the marker of terminal differentiation CD57, expand in response to HCMV. These NK-cell populations engaged in the long-lasting interaction with HCMV, in addition to characteristic but variable expression of surface receptors, exhibit reduced expression of signaling proteins and transcription factors expressed by canonical NK cells. Broad epigenetic modifications drive the emergence and persistence of HCMV-adapted NK cells that have distinct functional characteristics. NKG2C+ NK-cell expansions have been observed in HIV-1 infected patients and other acute and chronic viral infections being systematically associated with HCMV seropositivity. The latter is potentially an important confounding variable in studies focused on the cellular NK-cell receptor repertoire and functional capacity. Here, focusing on HIV-1 infection we review the evidence in favor of “adaptive” changes likely induced by HCMV co-infection in NK-cell subsets. We highlight a number of key questions and how insights into the adaptive behavior of NK cells will inform new strategies exploiting their unique properties in the fight against HIV-1.
Collapse
Affiliation(s)
- Dimitra Peppa
- Division of Infection and Immunity, University College London, London, United Kingdom.,Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
Falcão ASC, da Costa Vasconcelos PF, Lobato da Silva DDF, Viana Pinheiro JDJ, Falcão LFM, Quaresma JAS. Mechanisms of human cytomegalovirus infection with a focus on epidermal growth factor receptor interactions. Rev Med Virol 2017; 27. [PMID: 29024283 DOI: 10.1002/rmv.1955] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 09/01/2017] [Accepted: 09/18/2017] [Indexed: 12/21/2022]
Abstract
Human cytomegalovirus (HCMV) is a widespread opportunistic herpesvirus that causes severe diseases in immunocompromised individuals. It has a high prevalence worldwide that is linked with socioeconomic factors. Similar to other herpesviruses, HCMV has the ability to establish lifelong persistence and latent infection following primary exposure. HCMV infects a broad range of cell types. This broad tropism suggests that it may use multiple receptors for host cell entry. The identification of receptors used by HCMV is essential for understanding viral pathogenesis, because these receptors mediate the early events necessary for infection. Many cell surface components have been identified as virus receptors, such as epidermal growth factor receptor (EGFR), which is characterized by tyrosine kinase activity and plays a crucial role in the control of key cellular transduction pathways. EGFR is essential for HCMV binding, signaling, and host cell entry. This review focuses on HCMV infection via EGFR on different cell types and its implications for the cellular environment, viral persistence, and infection.
Collapse
Affiliation(s)
| | | | | | - João de Jesus Viana Pinheiro
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Federal University of Pará, Belém, Pará, Brazil.,Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Juarez Antonio Simões Quaresma
- Division of Infectious Diseases, Evandro Chagas Institute, Ananindeua, Pará, Brazil.,Center for Biological Sciences and Health, Pará State University, Belém, Pará, Brazil.,Division of Infectious Diseases, Tropical Medicine Center, Federal University of Pará, Belém, Pará, Brazil
| |
Collapse
|
46
|
van de Weijer ML, Schuren ABC, van den Boomen DJH, Mulder A, Claas FHJ, Lehner PJ, Lebbink RJ, Wiertz EJHJ. Multiple E2 ubiquitin-conjugating enzymes regulate human cytomegalovirus US2-mediated immunoreceptor downregulation. J Cell Sci 2017; 130:2883-2892. [PMID: 28743740 DOI: 10.1242/jcs.206839] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/10/2017] [Indexed: 12/13/2022] Open
Abstract
Misfolded endoplasmic reticulum (ER) proteins are dislocated towards the cytosol and degraded by the ubiquitin-proteasome system in a process called ER-associated protein degradation (ERAD). During infection with human cytomegalovirus (HCMV), the viral US2 protein targets HLA class I molecules (HLA-I) for degradation via ERAD to avoid elimination by the immune system. US2-mediated degradation of HLA-I serves as a paradigm of ERAD and has facilitated the identification of TRC8 (also known as RNF139) as an E3 ubiquitin ligase. No specific E2 enzymes had previously been described for cooperation with TRC8. In this study, we used a lentiviral CRISPR/Cas9 library targeting all known human E2 enzymes to assess their involvement in US2-mediated HLA-I downregulation. We identified multiple E2 enzymes involved in this process, of which UBE2G2 was crucial for the degradation of various immunoreceptors. UBE2J2, on the other hand, counteracted US2-induced ERAD by downregulating TRC8 expression. These findings indicate the complexity of cellular quality control mechanisms, which are elegantly exploited by HCMV to elude the immune system.
Collapse
Affiliation(s)
- Michael L van de Weijer
- Dept. Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Anouk B C Schuren
- Dept. Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | | | - Arend Mulder
- Dept. Immunohematology and blood transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Frans H J Claas
- Dept. Immunohematology and blood transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Paul J Lehner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Robert Jan Lebbink
- Dept. Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Emmanuel J H J Wiertz
- Dept. Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| |
Collapse
|
47
|
Abstract
Human cytomegalovirus (HCMV) encodes numerous proteins and microRNAs that function to evade the immune response and allow the virus to replicate and disseminate in the face of a competent innate and acquired immune system. The establishment of a latent infection by CMV, which if completely quiescent at the level of viral gene expression would represent an ultimate in immune evasion strategies, is not sufficient for lifelong persistence and dissemination of the virus. CMV needs to reactivate and replicate in a lytic cycle of infection in order to disseminate further, which occurs in the face of a fully primed secondary immune response. Without reactivation, latency itself would be redundant for the virus. It is also becoming clear that latency is not a totally quiescent state, but is characterized by limited viral gene expression. Therefore, the virus also needs immune evasion strategies during latency. An effective immune response to CMV is required or viral replication will cause morbidity and ultimately mortality in the host. There is clearly a complex balance between virus immune evasion and host immune recognition over a lifetime. This poses the important question of whether long-term evasion or manipulation of the immune response driven by CMV is detrimental to health. In this meeting report, three groups used the murine model of CMV (MCMV) to examine if the contribution of the virus to immune senescence is set by the (i) initial viral inoculum, (ii) inflation of T cell responses, (iii) or the balance between functionally distinct effector CD4+ T cells. The work of other groups studying the CMV response in humans is discussed. Their work asks whether the ability to make immune responses to new antigens is compromised by (i) age and HCMV carriage, (ii) long-term exposure to HCMV giving rise to an overall immunosuppressive environment and increased levels of latent virus, or (iii) adapted virus mutants (used as potential vaccines) that have the capacity to elicit conventional and unconventional T cell responses.
Collapse
|
48
|
Stangherlin LM, de Paula FN, Icimoto MY, Ruiz LGP, Nogueira ML, Braz ASK, Juliano L, da Silva MCC. Positively Selected Sites at HCMV gB Furin Processing Region and Their Effects in Cleavage Efficiency. Front Microbiol 2017; 8:934. [PMID: 28588572 PMCID: PMC5441137 DOI: 10.3389/fmicb.2017.00934] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 05/08/2017] [Indexed: 12/16/2022] Open
Abstract
Human cytomegalovirus is a ubiquitous infectious agent that affects mainly immunosuppressed, fetuses, and newborns. The virus has several polymorphic regions, in particular in the envelope glycoproteins. The UL55 gene encodes the glycoprotein B that has a variable region, containing a furin cleavage site and according to the variability different genotypes are characterized. Here we investigated variability and existence of selective pressure on the UL55 variable region containing the furin cleavage site in 213 clinical sequences from patients worldwide. We showed the occurrence of positive selective pressure on gB codons 461 and 462, near the furin cleavage site. Cleavage analysis of synthesized peptides demonstrated that most mutations confer better cleavage by furin, suggesting that evolution is acting in order to increase the efficiency cleavage and supporting the hypothesis that gB processing is important in the host. We also demonstrated that peptides containing sequences, that characterize genotypes gB2 and 3, are differentially cleaved by furin. Our data demonstrate for the first time that variability in the cleavage site is related to degree of gB processing by furin.
Collapse
Affiliation(s)
- Lucas M Stangherlin
- Center for Natural Sciences and Humanities, Federal University of ABCSanto André, Brazil
| | - Felipe N de Paula
- Center for Natural Sciences and Humanities, Federal University of ABCSanto André, Brazil.,Pasteur InstituteSão Paulo, Brazil
| | - Marcelo Y Icimoto
- Department of Biophysics, Paulista Medical School, Federal University of São PauloSão Paulo, Brazil
| | - Leonardo G P Ruiz
- Medical School of São José do Rio PretoSão José do Rio Preto, Brazil
| | | | - Antônio S K Braz
- Center for Natural Sciences and Humanities, Federal University of ABCSanto André, Brazil
| | - Luiz Juliano
- Department of Biophysics, Paulista Medical School, Federal University of São PauloSão Paulo, Brazil
| | - Maria C C da Silva
- Center for Natural Sciences and Humanities, Federal University of ABCSanto André, Brazil
| |
Collapse
|
49
|
Pollmann J, Rölle A, Hofmann M, Cerwenka A. Hepatitis C Virus and Human Cytomegalovirus-Natural Killer Cell Subsets in Persistent Viral Infections. Front Immunol 2017; 8:566. [PMID: 28567042 PMCID: PMC5434107 DOI: 10.3389/fimmu.2017.00566] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/27/2017] [Indexed: 12/17/2022] Open
Abstract
Hepatitis C virus (HCV) and human cytomegalovirus (HCMV) are prominent examples of RNA and DNA viruses, respectively, that establish a persistent infection in their host. HCV affects over 185 million patients worldwide, who are at high risk for developing liver fibrosis, liver cirrhosis, and ultimately hepatocellular carcinoma. Recent breakthroughs in HCV therapy, using direct-acting antivirals have provided the opportunity to monitor natural killer (NK) cells after clearance of a chronic infection. There is now increasing evidence that the individual NK cell repertoire before infection is predictive for the course of disease. HCMV affects the majority of the global population. While being asymptomatic in healthy individuals, HCMV represents a severe clinical challenge in immunocompromised patients. Both viral infections, HCV and HCMV, lead to long-lasting and profound alterations within the entire NK cell compartment. This review article, will discuss the diverse range of changes in the NK cell compartment as well as potential consequences for the course of disease.
Collapse
Affiliation(s)
- Julia Pollmann
- Research Group Innate Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Rölle
- Clinical Cooperation Unit Applied Tumor-Immunity, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Maike Hofmann
- Faculty of Medicine, Department of Medicine II, University Hospital Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | - Adelheid Cerwenka
- Research Group Innate Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical Faculty Mannheim, Division of Immunbiochemistry, University Heidelberg, Heidelberg, Germany
| |
Collapse
|
50
|
Kasztelewicz B, Czech-Kowalska J, Lipka B, Milewska-Bobula B, Borszewska-Kornacka MK, Romańska J, Dzierżanowska-Fangrat K. Cytokine gene polymorphism associations with congenital cytomegalovirus infection and sensorineural hearing loss. Eur J Clin Microbiol Infect Dis 2017; 36:1811-1818. [PMID: 28501927 PMCID: PMC5602083 DOI: 10.1007/s10096-017-2996-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/21/2017] [Indexed: 11/24/2022]
Abstract
Cytomegalovirus (CMV) is the most common viral agent of congenital infections and a leading nongenetic cause of sensorineural hearing loss (SNHL). The host immunologic factors that render a developing foetus prone to intrauterine CMV infection and development of hearing loss are unknown. The aim of this study was to assess the potential associations between the polymorphisms within cytokine and cytokine receptors genes, and the risk of congenital CMV infection, and the hearing outcome. A panel of 11 candidate single nucleotide polymorphisms (SNPs): TNF rs1799964, TNF rs1800629, TNFRSF1A rs4149570, IL1B rs16944, IL1B rs1143634, IL10 rs1800896, IL10RA rs4252279, IL12B rs3212227, CCL2 rs1024611, CCL2 rs13900, CCR5 rs333 was genotyped in 470 infants (72 with confirmed intrauterine CMV infection and 398 uninfected controls), and related to congenital CMV infection, and the outcome. In multivariate analysis, the IL1B rs16944 TT and TNF rs1799964 TC genotypes were significantly associated with intrauterine CMV infection (aOR = 2.32; 95% CI, 1.11–4.89; p = 0.032, and aOR = 2.17, 95% CI, 1.25–3.77; p = 0.007, respectively). Twenty-two out of 72 congenitally infected newborns had confirmed SNHL. Carriers of CT or TT genotype of CCL2 rs13900 had increased risk of hearing loss at birth and at 6 months of age (aOR = 3.59; p = 0.028 and aOR = 4.10; p = 0.039, respectively). This is the first study to report an association between SNPs in IL1B, TNF, and CCL2, and susceptibility to congenital CMV infection (IL1B and TNF) and SNHL (CCL2).
Collapse
Affiliation(s)
- B Kasztelewicz
- Department of Clinical Microbiology and Immunology, The Children's Memorial Health Institute, Dzieci Polskich 20, 04-730, Warsaw, Poland.
| | - J Czech-Kowalska
- Department of Neonatology and Neonatal Intensive Care, The Children's Memorial Health Institute, Warsaw, Poland
| | - B Lipka
- Department of Infant Diseases, The Children's Memorial Health Institute, Warsaw, Poland
| | - B Milewska-Bobula
- Department of Infant Diseases, The Children's Memorial Health Institute, Warsaw, Poland
| | | | - J Romańska
- Department of Neonatology, Warsaw Medical University Hospital, Warsaw, Poland
| | - K Dzierżanowska-Fangrat
- Department of Clinical Microbiology and Immunology, The Children's Memorial Health Institute, Dzieci Polskich 20, 04-730, Warsaw, Poland
| |
Collapse
|