1
|
Jeong H, Yeo N, Hwang H, Park J, Baek D, Ahn K. N6-methyladenosine modification of HCMV IE1 transcript promotes the repressive state of viral genome to achieve latent infection. Proc Natl Acad Sci U S A 2025; 122:e2508475122. [PMID: 40493198 DOI: 10.1073/pnas.2508475122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Accepted: 04/29/2025] [Indexed: 06/12/2025] Open
Abstract
Human cytomegalovirus (HCMV) is a prevalent pathogen that chronically infects the majority of human population. Among the many features that allow such widespread HCMV infection, one is its ability to maintain a transcriptionally dormant immune-evasive state called latency by suppressing its own major immediate early promoter (MIEP) via epigenetic alterations. In this study, we show a mechanism of MIEP regulation in which the major immediate early (MIE) gene product, immediate early 1 (IE1) transcript, downregulates its own promoter activity in an m6A modification-dependent manner. We found that the loss of the m6A writer, METTL3, in host cells impedes latency establishment in these cells. Through transcriptome-wide m6A profiling of latently infected monocytes, we identified that the major immediate early gene product IE1 transcript is m6A-modified during latent infection. Using IE1-specific m6A-abolished mutants, we found that m6A modification of the IE1 transcript was necessary for the efficient repression of MIEP, and these mutant viruses exhibited a significant defect in establishing latency and progressed toward lytic-like infection in the human monocytic cell line (THP-1) and primary CD14+ monocytes. Our findings demonstrate that HCMV exploits the host m6A machinery to suppress its own lytic program to establish latency and uncover an unexpected role of immediate early gene messenger RNA (mRNA) in regulating its own expression.
Collapse
Affiliation(s)
- Heena Jeong
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Republic of Korea
| | - Nagyeong Yeo
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyeonseo Hwang
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Junhyun Park
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Republic of Korea
| | - Daehyun Baek
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Kwangseog Ahn
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Republic of Korea
- Institute for Virus Research, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
2
|
Yi X, Liu L, Tao R, Li W, Li H, He L, Liu Y, Shang S. The effect of T cells on the lncRNA4.9-TGF-β1 axis in HCMV latently infected cells. Medicine (Baltimore) 2025; 104:e42400. [PMID: 40388758 PMCID: PMC12091603 DOI: 10.1097/md.0000000000042400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 04/22/2025] [Indexed: 05/21/2025] Open
Abstract
Human cytomegalovirus (HCMV), a prevalent double-stranded DNA virus, exhibits a high infectioraten, yet the mechanisms underlying latent infection and activation remain unclear. Viral cyclic reactivation in healthy HCMV and latent infection is usually well controlled by the T-cell response. Long noncoding RNA (lncRNA) is known to play vital roles in physiological and pathological processes. This study investigates the impact of T cells on the expression of lncRNA4.9, transforming growth factor-β (TGF-β), and multiple cytokines during HCMV latent infection. We established an HCMV latent infection model, coculturing human acute monocytic leukemia cell line (THP-1) cells with T cells subjected to different treatments: NC1 (THP-1 cells cocultured with untreated T cells), NC2 (HCMV latently infected group without T cells), phytohemagglutinin A (PHA) group (PHA-activated T cells added), FK506 group (FK506-suppressed T cells added), and T-cell group (untreated T cells added). Cytokines were assessed in cell culture supernatants collected at 24, 48, and 72 hours. Reverse transcription-quantitative polymerase chain reaction examined changes in RNA and HCMV DNA copy numbers after 3 and 5 days. In the HCMV latent infection model, PHA group, T-cell group, and FK506 group exhibited significantly increased interleukin (IL)-6, IL-10, and tumor necrosis factor-alpha secretion. Expressions of lncRNA4.9 and TGF-β1 significantly increased in T-cell group after 3 and 5 days. Expressions of lncRNA4.9 and TGF-β1 significantly decreased in the PHA group after 5 days. DNA copy numbers of HCMV decreased in T cell and PHA groups after 3 days, with no significant change after 5 days. This study reveals that PHA-activated T cells downregulate the expression of lncRNA4.9 and TGF-β1 in HCMV, highlighting the effect of T cells on the lncRNA4.9-TGF-β1 axis during HCMV latent infection. Regardless of T-cell activation, the study also indicates that IL-6, IL-10, and tumor necrosis factor-alpha levels increase during HCMV latent infection.
Collapse
Affiliation(s)
- Xiaolian Yi
- Department of Laboratory Center, Children’s Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Lifang Liu
- Department of Laboratory Center, Children’s Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Ran Tao
- Department of Laboratory Center, Children’s Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Li
- Department of Laboratory Center, Children’s Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Huamei Li
- Department of Laboratory Center, Children’s Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Lin He
- Department of Laboratory Center, Children’s Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Yujie Liu
- Department of Laboratory Center, Children’s Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Shiqiang Shang
- Department of Laboratory Center, Children’s Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Almeida GWC, Oliveira MT, Martines IGL, Fiori GC, Nevels MM, Groves IJ, Sinclair J, Medina-Pestana J, da Silva RS, Nakamura M, Requião-Moura L, Poole E, da Silva MCC. Expression Profile of Human Cytomegalovirus UL111A cmvIL-10 and LAcmvIL-10 Transcripts in Primary Cells and Cells from Renal Transplant Recipients. Viruses 2025; 17:501. [PMID: 40284944 PMCID: PMC12031159 DOI: 10.3390/v17040501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/11/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Human cytomegalovirus (HCMV) is a high-risk pathogen in immunocompromised individuals, especially in transplant recipients. HCMV viremia must be monitored, and frequently, patients are treated with antiviral agents. HCMV has a variety of strategies to modulate host antiviral responses, and one important player is a viral homolog of the cellular interleukin-10 (cIL-10). The viral UL111A gene produces several HCMV IL-10 transcripts and protein isoforms through alternative splicing. The cmvIL-10 (isoform A) has similar properties to cIL-10, while LAcmvIL-10 (isoform B) has more restricted biological properties. Other isoforms are produced (C to H) but have unknown functions. Here, we investigated the expression of the most abundant transcripts, cmvIL-10 and LAcmvIL-10, in productively and latently infected cells and in peripheral blood mononuclear cells from renal transplant recipients up to 60 days post-transplantation. This study investigated HCMV cmvIL-10 and LAcmvIL-10 transcription profiles in vitro, in productive and latent infection, and in vivo, in peripheral blood mononuclear cells (PBMCs) of renal transplant patients. In vitro, both cmvIL-10 and LAcmvIL-10 transcripts were detected in both types at high levels and low levels in MRC-5 and latent infected (CD14+). When PBMCs from transplant patients were analyzed, LAcmvIL-10 was detected mostly sporadically and in only a few patients, while cmvIL-10 was found in all patients at all time points. Furthermore, it was observed in PBMCs that expression of cmvIL-10 was positively associated with an increase in viral DNA detection in the subsequently collected sample, indicating that expression of cmvIL-10 might precede viral DNA replication. These results contribute to the understanding of HCMV biology in different phases of infection. In addition, our initial analysis suggests that monitoring cmvIL-10, along with viral DNA, could improve early detection of HCMV reactivation in transplant recipients.
Collapse
Affiliation(s)
- Giovana W. C. Almeida
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (G.W.C.A.); (M.T.O.); (I.G.L.M.); (G.C.F.)
| | - Martha T. Oliveira
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (G.W.C.A.); (M.T.O.); (I.G.L.M.); (G.C.F.)
| | - Isabella G. L. Martines
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (G.W.C.A.); (M.T.O.); (I.G.L.M.); (G.C.F.)
| | - Giuliano C. Fiori
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (G.W.C.A.); (M.T.O.); (I.G.L.M.); (G.C.F.)
| | - Michael M. Nevels
- School of Biology, University of St. Andrews, St. Andrews KY16 9ST, UK;
| | - Ian J. Groves
- Molecular Medicine, Cleveland Clinic, Cleveland, OH 44106, USA;
| | - John Sinclair
- Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK;
| | - José Medina-Pestana
- Hospital do Rim, Fundação Oswaldo Ramos, Vila Clementino 04038-002, SP, Brazil; (J.M.-P.); (R.S.d.S.); (M.N.); (L.R.-M.)
- Nephrology Division, Universidade Federal de São Paulo, Vila Clementino 04021-001, SP, Brazil
| | - Rayra Sampaio da Silva
- Hospital do Rim, Fundação Oswaldo Ramos, Vila Clementino 04038-002, SP, Brazil; (J.M.-P.); (R.S.d.S.); (M.N.); (L.R.-M.)
| | - Monica Nakamura
- Hospital do Rim, Fundação Oswaldo Ramos, Vila Clementino 04038-002, SP, Brazil; (J.M.-P.); (R.S.d.S.); (M.N.); (L.R.-M.)
| | - Lucio Requião-Moura
- Hospital do Rim, Fundação Oswaldo Ramos, Vila Clementino 04038-002, SP, Brazil; (J.M.-P.); (R.S.d.S.); (M.N.); (L.R.-M.)
- Nephrology Division, Universidade Federal de São Paulo, Vila Clementino 04021-001, SP, Brazil
| | - Emma Poole
- Department of Pathology, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Maria C. Carlan da Silva
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (G.W.C.A.); (M.T.O.); (I.G.L.M.); (G.C.F.)
| |
Collapse
|
4
|
Fares S, Krishna BA. Why Are Cytomegalovirus-Encoded G-Protein-Coupled Receptors Essential for Infection but Only Variably Conserved? Pathogens 2025; 14:245. [PMID: 40137730 PMCID: PMC11945030 DOI: 10.3390/pathogens14030245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Cytomegaloviruses (CMVs) encode viral G-protein-coupled receptors (vGPCRs) that have diverged from their cellular homologues to perform new functions. Human cytomegalovirus (HCMV) encodes four vGPCRs: UL33, UL78, US27, and US28, which contribute to viral pathogenesis, cellular signalling, and latency. While the role of US28 in chemokine signalling and viral latency is well characterised, the functions of other vGPCRs remain incompletely understood. Rodent cytomegaloviruses only have homologues to UL33 and UL78, while primates have two to five additional GPCRs which are homologues of US27 and US28. Different CMVs appear to have evolved vGPCRs with functions specific to infection of their respective host. As non-human CMVs are used as model organisms to understand clinical cytomegalovirus disease and develop vaccines and antivirals, understanding the differences between these vGPCRs helps researchers understand critical differences between their models. This review aims to address the differences between CMV vGPCRs, and how these differences may affect models of CMV disease to facilitate future research.
Collapse
Affiliation(s)
- Suzan Fares
- Occlutech Holding AG, Feldstrasse 22, 8200 Schaffhausen, Switzerland;
| | - Benjamin A. Krishna
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
5
|
Manska S, Hagemann A, Magana J, Rossetto CC, Verma SC. Characterization of Human Cytomegalovirus (HCMV) Long Non-Coding RNA1.2 During Lytic Replication. Viruses 2025; 17:149. [PMID: 40006904 PMCID: PMC11860937 DOI: 10.3390/v17020149] [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: 12/31/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 02/27/2025] Open
Abstract
During lytic replication of human cytomegalovirus (HCMV), the most abundant viral transcripts are long non-coding RNAs (lncRNAs). Viral lncRNAs can have a variety of functions, some of which are necessary for viral production and the modulation of host processes during infection. HCMV produces four lncRNAs, Beta2.7 (RNA2.7), RNA4.9, RNA5.0 and RNA1.2. While there has been research on these viral lncRNAs, many of their functions remain uncharacterized. To determine the function of RNA1.2, we explored its requirement during lytic infection by generating viral mutants containing either a full or partial deletion of the RNA1.2 locus. Within permissive fibroblasts, the RNA1.2 deletion mutants showed no defects in viral DNA synthesis, transcript expression, protein production, or generation of viral progeny. Further investigation to identify potential cellular and viral protein binding partners of RNA1.2 was performed using liquid chromatography-mass spectrometry (LC-MS). A significant number of cellular proteins were identified and associated with RNA1.2. Specifically those associated with the innate immune response, mitochondrial processes, and cell cycle regulation. While RNA1.2 is dispensable for lytic replication, these findings suggest it may play a pivotal role in modulating the host response.
Collapse
Affiliation(s)
| | | | | | | | - Subhash C. Verma
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA; (S.M.); (A.H.); (J.M.); (C.C.R.)
| |
Collapse
|
6
|
Payen SH, Adhikari K, Petereit J, Uppal T, Rossetto CC, Verma SC. SARS-CoV-2 superinfection in CD14 + monocytes with latent human cytomegalovirus (HCMV) promotes inflammatory cascade. Virus Res 2024; 345:199375. [PMID: 38642618 PMCID: PMC11061749 DOI: 10.1016/j.virusres.2024.199375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent of coronavirus disease 2019 (COVID-19), has posed significant challenges to global health. While much attention has been directed towards understanding the primary mechanisms of SARS-CoV-2 infection, emerging evidence suggests co-infections or superinfections with other viruses may contribute to increased morbidity and mortality, particularly in severe cases of COVID-19. Among viruses that have been reported in patients with SARS-CoV-2, seropositivity for Human cytomegalovirus (HCMV) is associated with increased COVID-19 risk and hospitalization. HCMV is a ubiquitous beta-herpesvirus with a seroprevalence of 60-90 % worldwide and one of the leading causes of mortality in immunocompromised individuals. The primary sites of latency for HCMV include CD14+ monocytes and CD34+ hematopoietic cells. In this study, we sought to investigate SARS-CoV-2 infection of CD14+ monocytes latently infected with HCMV. We demonstrate that CD14+ cells are susceptible and permissive to SARS-CoV-2 infection and detect subgenomic transcripts indicative of replication. To further investigate the molecular changes triggered by SARS-CoV-2 infection in HCMV-latent CD14+ monocytes, we conducted RNA sequencing coupled with bioinformatic differential gene analysis. The results revealed significant differences in cytokine-cytokine receptor interactions and inflammatory pathways in cells superinfected with replication-competent SARS-CoV-2 compared to the heat-inactivated and mock controls. Notably, there was a significant upregulation in transcripts associated with pro-inflammatory response factors and a decrease in anti-inflammatory factors. Taken together, these findings provide a basis for the heightened inflammatory response, offering potential avenues for targeted therapeutic interventions among HCMV-infected severe cases of COVID-19. SUMMARY: COVID-19 patients infected with secondary viruses have been associated with a higher prevalence of severe symptoms. Individuals seropositive for human cytomegalovirus (HCMV) infection are at an increased risk for severe COVID-19 disease and hospitalization. HCMV reactivation has been reported in severe COVID-19 cases with respiratory failure and could be the result of co-infection with SARS-CoV-2 and HCMV. In a cell culture model of superinfection, HCMV has previously been shown to increase infection of SARS-CoV-2 of epithelial cells by upregulating the human angiotensin-converting enzyme-2 (ACE2) receptor. In this study, we utilize CD14+ monocytes, a major cell type that harbors latent HCMV, to investigate co-infection of SARS-CoV-2 and HCMV. This study is a first step toward understanding the mechanism that may facilitate increased COVID-19 disease severity in patients infected with SARS-CoV-2 and HCMV.
Collapse
Affiliation(s)
- Shannon Harger Payen
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Kabita Adhikari
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Juli Petereit
- Nevada Bioinformatics Center (RRID:SCR_017802), University of Nevada, Reno, NV 89557, United States
| | - Timsy Uppal
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Cyprian C Rossetto
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Subhash C Verma
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States.
| |
Collapse
|
7
|
Guyon J, Haidar Ahmad S, El Baba R, Le Quang M, Bikfalvi A, Daubon T, Herbein G. Generation of glioblastoma in mice engrafted with human cytomegalovirus-infected astrocytes. Cancer Gene Ther 2024; 31:1070-1080. [PMID: 38553638 PMCID: PMC11257955 DOI: 10.1038/s41417-024-00767-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 07/20/2024]
Abstract
Mounting evidence is identifying human cytomegalovirus (HCMV) as a potential oncogenic virus. HCMV has been detected in glioblastoma multiforme (GB). Herewith, we present the first experimental evidence for the generation of CMV-Elicited Glioblastoma Cells (CEGBCs) possessing glioblastoma-like traits that lead to the formation of glioblastoma in orthotopically xenografted mice. In addition to the already reported oncogenic HCMV-DB strain, we isolated three HCMV clinical strains from GB tissues that transformed HAs toward CEGBCs and generated spheroids from CEGBCs that resulted in the appearance of glioblastoma-like tumors in xenografted mice. These tumors were nestin-positive mostly in the invasive part surrounded by GFAP-positive reactive astrocytes. The glioblastoma immunohistochemistry phenotype was confirmed by EGFR and cMet gene amplification in the tumor parallel to the detection of HCMV IE and UL69 genes and proteins. Our results fit with an HCMV-induced glioblastoma model of oncogenesis in vivo which will open the door to new therapeutic approaches and assess the anti-HCMV treatment as well as immunotherapy in fighting GB which is characterized by poor prognosis.
Collapse
Affiliation(s)
- Joris Guyon
- University of Bordeaux, INSERM U1312, BRIC, Bordeaux, France
- CHU Bordeaux, Department of Medical Pharmacology, Bordeaux, France
| | - Sandy Haidar Ahmad
- University of Franche-Comté, Pathogens & Inflammation/EPILAB Laboratory, EA 4266, Besançon, France
| | - Ranim El Baba
- University of Franche-Comté, Pathogens & Inflammation/EPILAB Laboratory, EA 4266, Besançon, France
| | - Mégane Le Quang
- Pathology Department, University Hospital of Bordeaux, Bordeaux, France
| | | | - Thomas Daubon
- University of Bordeaux, CNRS, IBGC UMR5095, Bordeaux, France
| | - Georges Herbein
- University of Franche-Comté, Pathogens & Inflammation/EPILAB Laboratory, EA 4266, Besançon, France.
- CHU Besançon, Department of Virology, Besançon, France.
| |
Collapse
|
8
|
Bradley E, Poole E, Reeves MB. The Triterpenoid MOMORDIN-Ic Inhibits HCMV by Preventing the Initiation of Gene Expression in Eukaryotic Cells. Pathogens 2024; 13:546. [PMID: 39057773 PMCID: PMC11280373 DOI: 10.3390/pathogens13070546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Human cytomegalovirus (HCMV) primary infection, re-infection, and reactivation from latency cause morbidity in immune-compromised patients. Consequently, potential therapeutic strategies remain of interest for the treatment of infection. Naturally occurring triterpenoids derived from plants have been demonstrated to have anti-viral activity, although their precise mechanisms of action are not always fully understood. Here, we investigate the activity of Mormordin Ic (Mc) and demonstrate that it is potently anti-viral against HCMV. Through investigation of the mechanistic basis of this anti-viral activity, we identify that it is inhibitory to both viral and host gene expression, and to highly induced genes in particular. We go on to observe that Mc impacts on RNA Pol II activity and, specifically, reduces the occupancy of elongating RNA Pol II at a viral promoter. Next, we demonstrate that Mc is inhibitory to HCMV reactivation, and in doing so identify that it has greater activity against the canonical major immediate early promoter compared to the alternative ip2 promoter located downstream. Finally, we see evidence of RNA Pol II occupancy at the ip2 promoter in undifferentiated myeloid cells. Thus, Mc is potently anti-viral and a potential tool to probe the activity of multiple promoters considered important for controlling HCMV reactivation.
Collapse
Affiliation(s)
- Eleanor Bradley
- Institute of Immunity & Transplantation, Division of Infection & Immunity, UCL, Royal Free Campus, London NW3 2PP, UK;
| | - Emma Poole
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke’s Campus, Cambridge CB2 0QQ, UK;
| | - Matthew B. Reeves
- Institute of Immunity & Transplantation, Division of Infection & Immunity, UCL, Royal Free Campus, London NW3 2PP, UK;
| |
Collapse
|
9
|
Collins-McMillen D, De Oliveira Pessoa D, Zarrella K, Parkins CJ, Daily M, Moorman NJ, Kamil JP, Caposio P, Padi M, Goodrum FD. Viral and host network analysis of the human cytomegalovirus transcriptome in latency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.594597. [PMID: 38826434 PMCID: PMC11142044 DOI: 10.1101/2024.05.21.594597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
HCMV genes UL135 and UL138 play opposing roles regulating latency and reactivation in CD34+ human progenitor cells (HPCs). Using the THP-1 cell line model for latency and reactivation, we designed an RNA sequencing study to compare the transcriptional profile of HCMV infection in the presence and absence of these genes. The loss of UL138 results in elevated levels of viral gene expression and increased differentiation of cell populations that support HCMV gene expression and genome synthesis. The loss of UL135 results in diminished viral gene expression during an initial burst that occurs as latency is established and no expression of eleven viral genes from the ULb' region even following stimulation for differentiation and reactivation. Transcriptional network analysis revealed host transcription factors with potential to regulate the ULb' genes in coordination with pUL135. These results reveal roles for UL135 and UL138 in regulation of viral gene expression and potentially hematopoietic differentiation.
Collapse
Affiliation(s)
- Donna Collins-McMillen
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Diogo De Oliveira Pessoa
- Bioinformatics Shared Resource, Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| | - Kristen Zarrella
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Christopher J. Parkins
- Vaccine and Gene Therapy Institute, Oregon Health Science University, Beaverton, Oregon, United States of America
| | - Michael Daily
- Vaccine and Gene Therapy Institute, Oregon Health Science University, Beaverton, Oregon, United States of America
| | - Nathaniel J. Moorman
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jeremy P. Kamil
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health Science University, Beaverton, Oregon, United States of America
| | - Megha Padi
- Bioinformatics Shared Resource, Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| | - Felicia D. Goodrum
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
10
|
Costa B, Becker J, Krammer T, Mulenge F, Durán V, Pavlou A, Gern OL, Chu X, Li Y, Čičin-Šain L, Eiz-Vesper B, Messerle M, Dölken L, Saliba AE, Erhard F, Kalinke U. Human cytomegalovirus exploits STING signaling and counteracts IFN/ISG induction to facilitate infection of dendritic cells. Nat Commun 2024; 15:1745. [PMID: 38409141 PMCID: PMC10897438 DOI: 10.1038/s41467-024-45614-3] [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: 10/01/2021] [Accepted: 01/30/2024] [Indexed: 02/28/2024] Open
Abstract
Human cytomegalovirus (HCMV) is a widespread pathogen that in immunocompromised hosts can cause life-threatening disease. Studying HCMV-exposed monocyte-derived dendritic cells by single-cell RNA sequencing, we observe that most cells are entered by the virus, whereas less than 30% of them initiate viral gene expression. Increased viral gene expression is associated with activation of the stimulator of interferon genes (STING) that usually induces anti-viral interferon responses, and with the induction of several pro- (RHOB, HSP1A1, DNAJB1) and anti-viral (RNF213, TNFSF10, IFI16) genes. Upon progression of infection, interferon-beta but not interferon-lambda transcription is inhibited. Similarly, interferon-stimulated gene expression is initially induced and then shut off, thus further promoting productive infection. Monocyte-derived dendritic cells are composed of 3 subsets, with one being especially susceptible to HCMV. In conclusion, HCMV permissiveness of monocyte-derived dendritic cells depends on complex interactions between virus sensing, regulation of the interferon response, and viral gene expression.
Collapse
Grants
- 158989968 - SFB 900-B2 Deutsche Forschungsgemeinschaft (German Research Foundation)
- 398367752 - FOR 2830 Deutsche Forschungsgemeinschaft (German Research Foundation)
- EXC 2155 "RESIST" - Project ID 39087428 Deutsche Forschungsgemeinschaft (German Research Foundation)
- DO 1275/7-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- ER 927/2-1 - FOR2830 Deutsche Forschungsgemeinschaft (German Research Foundation)
- COALITION Niedersächsisches Ministerium für Wissenschaft und Kultur (Ministry for Science and Culture of Lower Saxony)
- Marie Skłodowska-Curie Actions Innovative Training Network (VIROINF: 955974) European Commission (EC)
- Marie Skłodowska-Curie Actions Innovative Training Network (VIROINF: 955974) European Commission (EC)
- 0703/68674/5/2017 Bayerisches Staatsministerium für Wirtschaft und Medien, Energie und Technologie (Bavarian Ministry of Economic Affairs and Media, Energy and Technology)
- 0703/89374/3/2017 Bayerisches Staatsministerium für Wirtschaft und Medien, Energie und Technologie (Bavarian Ministry of Economic Affairs and Media, Energy and Technology)
- 0703/68674/5/2017 Bayerisches Staatsministerium für Wirtschaft und Medien, Energie und Technologie (Bavarian Ministry of Economic Affairs and Media, Energy and Technology)
- 0703/89374/3/2017 Bayerisches Staatsministerium für Wirtschaft und Medien, Energie und Technologie (Bavarian Ministry of Economic Affairs and Media, Energy and Technology)
- 0703/68674/5/2017 Bayerisches Staatsministerium für Wirtschaft und Medien, Energie und Technologie (Bavarian Ministry of Economic Affairs and Media, Energy and Technology)
- 0703/89374/3/2017 Bayerisches Staatsministerium für Wirtschaft und Medien, Energie und Technologie (Bavarian Ministry of Economic Affairs and Media, Energy and Technology)
Collapse
Affiliation(s)
- Bibiana Costa
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Jennifer Becker
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Tobias Krammer
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Centre for Infection Research (HZI), Würzburg, Germany
| | - Felix Mulenge
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Verónica Durán
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Andreas Pavlou
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Olivia Luise Gern
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Xiaojing Chu
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM) & TWINCORE, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Yang Li
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM) & TWINCORE, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Luka Čičin-Šain
- Institute for Immune Aging and Chronic Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Britta Eiz-Vesper
- Institute for Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Martin Messerle
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Lars Dölken
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Centre for Infection Research (HZI), Würzburg, Germany
- University of Würzburg, Faculty of Medicine, Institute of Molecular Infection Biology (IMIB), Würzburg, Germany
| | - Florian Erhard
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany.
- Faculty for Informatics and Data Science, University of Regensburg, Regensburg, Germany.
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany.
- Cluster of Excellence - Resolving Infection Susceptibility (RESIST, EXC 2155), Hannover Medical School, Hannover, Germany.
| |
Collapse
|
11
|
Deng Y, Ostermann E, Brune W. A cytomegalovirus inflammasome inhibitor reduces proinflammatory cytokine release and pyroptosis. Nat Commun 2024; 15:786. [PMID: 38278864 PMCID: PMC10817922 DOI: 10.1038/s41467-024-45151-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
In response to viral infection, cells can initiate programmed cell death (PCD), leading to a reduction in the release of viral progeny. Viruses have therefore evolved specific mechanisms to curb PCD. Cytomegaloviruses (CMVs) are sophisticated manipulators of cellular defenses and encode potent inhibitors of apoptosis and necroptosis. However, a CMV inhibitor of pyroptosis has not been clearly identified and characterized. Here we identify the mouse cytomegalovirus M84 protein as an inhibitor of pyroptosis and proinflammatory cytokine release. M84 interacts with the pyrin domain of AIM2 and ASC to inhibit inflammasome assembly. It thereby prevents Caspase-1-mediated activation of interleukin 1β (IL-1β), IL-18, and Gasdermin D. Growth attenuation of an M84-deficient MCMV in macrophages is rescued by knockout of either Aim2 or Asc or by treatment with a Caspase-1 inhibitor, and its attenuation in infected mice is partially rescued in Asc knockout mice. Thus, viral inhibition of the inflammasome-pyroptosis pathway is important to promote viral replication in vivo.
Collapse
Affiliation(s)
- Yingqi Deng
- Leibniz Institute of Virology (LIV), Hamburg, Germany
| | | | - Wolfram Brune
- Leibniz Institute of Virology (LIV), Hamburg, Germany.
| |
Collapse
|
12
|
Torma G, Tombácz D, Csabai Z, Almsarrhad IAA, Nagy GÁ, Kakuk B, Gulyás G, Spires LM, Gupta I, Fülöp Á, Dörmő Á, Prazsák I, Mizik M, Dani VÉ, Csányi V, Harangozó Á, Zádori Z, Toth Z, Boldogkői Z. Identification of herpesvirus transcripts from genomic regions around the replication origins. Sci Rep 2023; 13:16395. [PMID: 37773348 PMCID: PMC10541914 DOI: 10.1038/s41598-023-43344-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023] Open
Abstract
Long-read sequencing (LRS) techniques enable the identification of full-length RNA molecules in a single run eliminating the need for additional assembly steps. LRS research has exposed unanticipated transcriptomic complexity in various organisms, including viruses. Herpesviruses are known to produce a range of transcripts, either close to or overlapping replication origins (Oris) and neighboring genes related to transcription or replication, which possess confirmed or potential regulatory roles. In our research, we employed both new and previously published LRS and short-read sequencing datasets to uncover additional Ori-proximal transcripts in nine herpesviruses from all three subfamilies (alpha, beta and gamma). We discovered novel long non-coding RNAs, as well as splice and length isoforms of mRNAs. Moreover, our analysis uncovered an intricate network of transcriptional overlaps within the examined genomic regions. We demonstrated that herpesviruses display distinct patterns of transcriptional overlaps in the vicinity of or at the Oris. Our findings suggest the existence of a 'super regulatory center' in the genome of alphaherpesviruses that governs the initiation of both DNA replication and global transcription through multilayered interactions among the molecular machineries.
Collapse
Affiliation(s)
- Gábor Torma
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Dóra Tombácz
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- MTA -SZTE Lendület GeMiNI Research Group, University of Szeged, Szeged, Hungary
| | - Zsolt Csabai
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- MTA -SZTE Lendület GeMiNI Research Group, University of Szeged, Szeged, Hungary
| | - Islam A A Almsarrhad
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Gergely Ármin Nagy
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Balázs Kakuk
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- MTA -SZTE Lendület GeMiNI Research Group, University of Szeged, Szeged, Hungary
| | - Gábor Gulyás
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- MTA -SZTE Lendület GeMiNI Research Group, University of Szeged, Szeged, Hungary
| | - Lauren McKenzie Spires
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Ishaan Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi, India
| | - Ádám Fülöp
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Ákos Dörmő
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- MTA -SZTE Lendület GeMiNI Research Group, University of Szeged, Szeged, Hungary
| | - István Prazsák
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- MTA -SZTE Lendület GeMiNI Research Group, University of Szeged, Szeged, Hungary
| | - Máté Mizik
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Virág Éva Dani
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Viktor Csányi
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Ákos Harangozó
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Zoltán Zádori
- HUN-REN Veterinary Medical Research Institute HU, Budapest, Hungary
| | - Zsolt Toth
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Zsolt Boldogkői
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
- MTA -SZTE Lendület GeMiNI Research Group, University of Szeged, Szeged, Hungary.
| |
Collapse
|
13
|
Gorbea C, Elhakiem A, Cazalla D. Shaping the host cell environment with viral noncoding RNAs. Semin Cell Dev Biol 2023; 146:20-30. [PMID: 36581481 PMCID: PMC10101873 DOI: 10.1016/j.semcdb.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/24/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022]
Abstract
Just like the cells they infect viruses express different classes of noncoding RNAs (ncRNAs). Viral ncRNAs come in all shapes and forms, and they usually associate with cellular proteins that are important for their functions. Viral ncRNAs have diverse functions, but they all contribute to the viral control of the cellular environment. Viruses utilize ncRNAs to regulate viral replication, to decide whether they should remain latent or reactivate, to evade the host immune responses, or to promote cellular transformation. In this review we describe the diverse functions played by different classes of ncRNAs expressed by adenoviruses and herpesviruses, how they contribute to the viral infection, and how their study led to insights into RNA-based mechanisms at play in host cells.
Collapse
Affiliation(s)
- Carlos Gorbea
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Abdalla Elhakiem
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Demián Cazalla
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
14
|
Cheung J, Remiszewski S, Chiang LW, Ahmad E, Pal M, Rahman SA, Nikolovska-Coleska Z, Chan GC. Inhibition of SIRT2 promotes death of human cytomegalovirus-infected peripheral blood monocytes via apoptosis and necroptosis. Antiviral Res 2023; 217:105698. [PMID: 37562606 DOI: 10.1016/j.antiviral.2023.105698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023]
Abstract
Peripheral blood monocytes are the cells predominantly responsible for systemic dissemination of human cytomegalovirus (HCMV) and a significant cause of morbidity and mortality in immunocompromised patients. HCMV establishes a silent/quiescent infection in monocytes, which is defined by the lack of viral replication and lytic gene expression. The absence of replication shields the virus within infected monocytes from the current available antiviral drugs that are designed to suppress active replication. Our previous work has shown that HCMV stimulates a noncanonical phosphorylation of Akt and the subsequent upregulation of a distinct subset of prosurvival proteins in normally short-lived monocytes. In this study, we found that SIRT2 activity is required for the unique activation profile of Akt induced within HCMV-infected monocytes. Importantly, both therapeutic and prophylactic treatment with a novel SIRT2 inhibitor, FLS-379, promoted death of infected monocytes via both the apoptotic and necroptotic cell death pathways. Mechanistically, SIRT2 inhibition reduced expression of Mcl-1, an Akt-dependent antiapoptotic Bcl-2 family member, and enhanced activation of MLKL, the executioner kinase of necroptosis. We have previously reported HCMV to block necroptosis by stimulating cellular autophagy. Here, we additionally demonstrate that inhibition of SIRT2 suppressed Akt-dependent HCMV-induced autophagy leading to necroptosis of infected monocytes. Overall, our data show that SIRT2 inhibition can simultaneously promote death of quiescently infected monocytes by two distinct death pathways, apoptosis and necroptosis, which may be vital for limiting viral dissemination to peripheral organs in immunosuppressed patients.
Collapse
Affiliation(s)
- Jennifer Cheung
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Stacy Remiszewski
- Evrys Bio, LLC, Pennsylvania Biotechnology Center, Doylestown, PA, 18902, USA
| | - Lillian W Chiang
- Evrys Bio, LLC, Pennsylvania Biotechnology Center, Doylestown, PA, 18902, USA
| | - Ejaz Ahmad
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Mohan Pal
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Sm Ashikur Rahman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Zaneta Nikolovska-Coleska
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gary C Chan
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
15
|
Abstract
Human cytomegalovirus (HCMV) is a betaherpesvirus that establishes lifelong infection in its host and can cause severe comorbidities in individuals with suppressed or compromised immune systems. The lifecycle of HCMV consists of lytic and latent phases, largely dependent upon the cell type infected and whether transcription from the major immediate early locus can ensue. Control of this locus, which acts as a critical "switch" region from where the lytic gene expression cascade originates, as well as regulation of the additional ~235 kilobases of virus genome, occurs through chromatinization with cellular histone proteins after infection. Upon infection of a host cell, an initial intrinsic antiviral response represses gene expression from the incoming genome, which is relieved in permissive cells by viral and host factors in concert. Latency is established in a subset of hematopoietic cells, during which viral transcription is largely repressed while the genome is maintained. As these latently infected cells differentiate, the cellular milieu and epigenetic modifications change, giving rise to the initial stages of virus reactivation from latency. Thus, throughout the cycle of infection, chromatinization, chromatin modifiers, and the recruitment of specific transcription factors influence the expression of genes from the HCMV genome. In this review, we discuss epigenetic regulation of the HCMV genome during the different phases of infection, with an emphasis on recent reports that add to our current perspective.
Collapse
Affiliation(s)
- Stephen M. Matthews
- Infection Biology, Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ian J. Groves
- Infection Biology, Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic, Cleveland, Ohio, USA
| | - Christine M. O'Connor
- Infection Biology, Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
16
|
Zeng J, Cao D, Yang S, Jaijyan DK, Liu X, Wu S, Cruz-Cosme R, Tang Q, Zhu H. Insights into the Transcriptome of Human Cytomegalovirus: A Comprehensive Review. Viruses 2023; 15:1703. [PMID: 37632045 PMCID: PMC10458407 DOI: 10.3390/v15081703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a widespread pathogen that poses significant risks to immunocompromised individuals. Its genome spans over 230 kbp and potentially encodes over 200 open-reading frames. The HCMV transcriptome consists of various types of RNAs, including messenger RNAs (mRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and microRNAs (miRNAs), with emerging insights into their biological functions. HCMV mRNAs are involved in crucial viral processes, such as viral replication, transcription, and translation regulation, as well as immune modulation and other effects on host cells. Additionally, four lncRNAs (RNA1.2, RNA2.7, RNA4.9, and RNA5.0) have been identified in HCMV, which play important roles in lytic replication like bypassing acute antiviral responses, promoting cell movement and viral spread, and maintaining HCMV latency. CircRNAs have gained attention for their important and diverse biological functions, including association with different diseases, acting as microRNA sponges, regulating parental gene expression, and serving as translation templates. Remarkably, HCMV encodes miRNAs which play critical roles in silencing human genes and other functions. This review gives an overview of human cytomegalovirus and current research on the HCMV transcriptome during lytic and latent infection.
Collapse
Affiliation(s)
- Janine Zeng
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA
| | - Di Cao
- Department of Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518052, China
| | - Shaomin Yang
- Department of Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518052, China
| | - Dabbu Kumar Jaijyan
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA
| | - Xiaolian Liu
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Songbin Wu
- Department of Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518052, China
| | - Ruth Cruz-Cosme
- Department of Microbiology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| | - Hua Zhu
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA
| |
Collapse
|
17
|
Crawford LB. Hematopoietic stem cells and betaherpesvirus latency. Front Cell Infect Microbiol 2023; 13:1189805. [PMID: 37346032 PMCID: PMC10279960 DOI: 10.3389/fcimb.2023.1189805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
The human betaherpesviruses including human cytomegalovirus (HCMV), human herpesvirus (HHV)-6a and HHV-6b, and HHV-7 infect and establish latency in CD34+ hematopoietic stem and progenitor cells (HPCs). The diverse repertoire of HPCs in humans and the complex interactions between these viruses and host HPCs regulate the viral lifecycle, including latency. Precise manipulation of host and viral factors contribute to preferential maintenance of the viral genome, increased host cell survival, and specific manipulation of the cellular environment including suppression of neighboring cells and immune control. The dynamic control of these processes by the virus regulate inter- and intra-host signals critical to the establishment of chronic infection. Regulation occurs through direct viral protein interactions and cellular signaling, miRNA regulation, and viral mimics of cellular receptors and ligands, all leading to control of cell proliferation, survival, and differentiation. Hematopoietic stem cells have unique biological properties and the tandem control of virus and host make this a unique environment for chronic herpesvirus infection in the bone marrow. This review highlights the elegant complexities of the betaherpesvirus latency and HPC virus-host interactions.
Collapse
Affiliation(s)
- Lindsey B Crawford
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
18
|
Ijezie EC, O'Dowd JM, Kuan MI, Faeth AR, Fortunato EA. HCMV Infection Reduces Nidogen-1 Expression, Contributing to Impaired Neural Rosette Development in Brain Organoids. J Virol 2023; 97:e0171822. [PMID: 37125912 PMCID: PMC10231252 DOI: 10.1128/jvi.01718-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a leading cause of birth defects in humans. These birth defects include microcephaly, sensorineural hearing loss, vision loss, and cognitive impairment. The process by which the developing fetus incurs these neurological defects is poorly understood. To elucidate some of these mechanisms, we have utilized HCMV-infected induced pluripotent stem cells (iPSCs) to generate in vitro brain organoids, modeling the first trimester of fetal brain development. Early during culturing, brain organoids generate neural rosettes. These structures are believed to model neural tube formation. Rosette formation was analyzed in HCMV-infected and mock-infected brain organoids at 17, 24, and 31 days postinfection. Histological analysis revealed fewer neural rosettes in HCMV-infected compared to mock-infected organoids. HCMV-infected organoid rosettes incurred multiple structural deficits, including increased lumen area, decreased ventricular zone depth, and decreased cell count. Immunofluorescent (IF) analysis found that nidogen-1 (NID1) protein expression in the basement membrane surrounding neural rosettes was greatly reduced by virus infection. IF analysis also identified a similar downregulation of laminin in basement membranes of HCMV-infected organoid rosettes. Knockdown of NID1 alone in brain organoids impaired their development, leading to the production of rosettes with increased lumen area, decreased structural integrity, and reduced laminin localization in the basement membrane, paralleling observations in HCMV-infected organoids. Our data strongly suggest that HCMV-induced downregulation of NID1 impairs neural rosette formation and integrity, likely contributing to many of HCMV's most severe birth defects. IMPORTANCE HCMV infection in pregnant women continues to be the leading cause of virus-induced neurologic birth defects. The mechanism through which congenital HCMV (cCMV) infection induces pathological changes to the developing fetal central nervous system (CNS) remains unclear. Our lab previously reproduced identified clinical defects in HCMV-infected infants using a three dimensional (3D) brain organoid model. In this new study, we have striven to discover very early HCMV-induced changes in developing brain organoids. We investigated the development of neural tube-like structures, neural rosettes. HCMV-infected rosettes displayed multiple structural abnormalities and cell loss. HCMV-infected rosettes displayed reduced expression of the key basement membrane protein, NID1. We previously found NID1 to be specifically targeted in HCMV-infected fibroblasts and endothelial cells. Brain organoids generated from NID1 knockdown iPSCs recapitulated the structural defects observed in HCMV-infected rosettes. Findings in this study revealed HCMV infection induced early and dramatic structural changes in 3D brain organoids. We believe our results suggest a major role for infection-induced NID1 downregulation in HCMV-induced CNS birth defects.
Collapse
Affiliation(s)
- Emmanuel C. Ijezie
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, Idaho, USA
| | - John M. O'Dowd
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, Idaho, USA
| | - Man I Kuan
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, Idaho, USA
| | - Alexandra R. Faeth
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, Idaho, USA
| | - Elizabeth A. Fortunato
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, Idaho, USA
| |
Collapse
|
19
|
El Baba R, Pasquereau S, Haidar Ahmad S, Monnien F, Abad M, Bibeau F, Herbein G. EZH2-Myc driven glioblastoma elicited by cytomegalovirus infection of human astrocytes. Oncogene 2023:10.1038/s41388-023-02709-3. [PMID: 37147437 DOI: 10.1038/s41388-023-02709-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/07/2023]
Abstract
Mounting evidence is identifying human cytomegalovirus (HCMV) as a potential oncogenic virus. HCMV has been detected in malignant gliomas. EZH2 and Myc play a potential oncogenic role, correlating with the glioma grade. Herewith, we present the first experimental evidence for HCMV as a reprogramming vector, straight through the dedifferentiation of mature human astrocytes, and generation of CMV-Elicited Glioblastoma Cells (CEGBCs) possessing glioblastoma-like traits. HCMV counterparts the progression of the perceived cellular and molecular mechanisms succeeding the transformation and invasion processes with CEGBCs involved in spheroid formation and invasiveness. Glioblastoma multiforme (GBM) biopsies were characterized by an elevated EZH2 and Myc expression, possessing a strong positive correlation between the aforementioned markers in the presence of HCMV. From GBM tissues, we isolated HCMV clinical strains that transformed HAs toward CEGBCs exhibiting upregulated EZH2 and Myc. Spheroids generated from CEGBCs possessed invasion potential and were sensitive to EZH2 inhibitor, ganciclovir, and temozolomide triple therapy. HCMV clinical strains transform HAs and fit with an HCMV-induced glioblastoma model of oncogenesis, and supports the tumorigenic properties of Myc and EZH2 which might be highly pertinent in the pathophysiology of astrocytic brain tumors and thereby paving the way for new therapeutic strategies.
Collapse
Affiliation(s)
- Ranim El Baba
- Department of Pathogens & Inflammation-EPILAB Laboratory EA4266, University of Franche-Comté, Besançon, France
| | - Sébastien Pasquereau
- Department of Pathogens & Inflammation-EPILAB Laboratory EA4266, University of Franche-Comté, Besançon, France
| | - Sandy Haidar Ahmad
- Department of Pathogens & Inflammation-EPILAB Laboratory EA4266, University of Franche-Comté, Besançon, France
| | | | - Marine Abad
- Department of Pathology, CHU Besançon, Besançon, France
| | | | - Georges Herbein
- Department of Pathogens & Inflammation-EPILAB Laboratory EA4266, University of Franche-Comté, Besançon, France.
- Department of Virology, CHU Besançon, Besançon, France.
| |
Collapse
|
20
|
Kulkarni V, Jayakumar S, Mohan M, Kulkarni S. Aid or Antagonize: Nuclear Long Noncoding RNAs Regulate Host Responses and Outcomes of Viral Infections. Cells 2023; 12:987. [PMID: 37048060 PMCID: PMC10093752 DOI: 10.3390/cells12070987] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are transcripts measuring >200 bp in length and devoid of protein-coding potential. LncRNAs exceed the number of protein-coding mRNAs and regulate cellular, developmental, and immune pathways through diverse molecular mechanisms. In recent years, lncRNAs have emerged as epigenetic regulators with prominent roles in health and disease. Many lncRNAs, either host or virus-encoded, have been implicated in critical cellular defense processes, such as cytokine and antiviral gene expression, the regulation of cell signaling pathways, and the activation of transcription factors. In addition, cellular and viral lncRNAs regulate virus gene expression. Viral infections and associated immune responses alter the expression of host lncRNAs regulating immune responses, host metabolism, and viral replication. The influence of lncRNAs on the pathogenesis and outcomes of viral infections is being widely explored because virus-induced lncRNAs can serve as diagnostic and therapeutic targets. Future studies should focus on thoroughly characterizing lncRNA expressions in virus-infected primary cells, investigating their role in disease prognosis, and developing biologically relevant animal or organoid models to determine their suitability for specific therapeutic targeting. Many cellular and viral lncRNAs localize in the nucleus and epigenetically modulate viral transcription, latency, and host responses to infection. In this review, we provide an overview of the role of nuclear lncRNAs in the pathogenesis and outcomes of viral infections, such as the Influenza A virus, Sendai Virus, Respiratory Syncytial Virus, Hepatitis C virus, Human Immunodeficiency Virus, and Herpes Simplex Virus. We also address significant advances and barriers in characterizing lncRNA function and explore the potential of lncRNAs as therapeutic targets.
Collapse
Affiliation(s)
- Viraj Kulkarni
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA;
| | - Sahana Jayakumar
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (S.J.); (M.M.)
| | - Mahesh Mohan
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (S.J.); (M.M.)
| | - Smita Kulkarni
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (S.J.); (M.M.)
| |
Collapse
|
21
|
Zhao F, Fung TY, Chen Z, Wang H, Cheung AKL. Association of human cytomegalovirus in urine with end-organ diseases in stage 2/3 HIV-1-infected individuals. J Clin Virol 2023; 158:105351. [PMID: 36529059 DOI: 10.1016/j.jcv.2022.105351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Human cytomegalovirus (HCMV) is prevalent in human immunodeficiency virus type 1 (HIV-1)-infected individuals but is suppressed by the host immune system bolstered by antiretroviral therapy. During stage 4 of HIV-1 infection, HCMV becomes a major risk factor for end-organ diseases (EODs). However, the implications of detecting HCMV in patients with stage 2/3 HIV-1 infection have not been established. OBJECTIVES Conduct a retrospective study of the relationship between HCMV-DNA detection and EODs in patients with stage 2/3 HIV-1 infection. STUDY DESIGN We cross-sectionally analyzed data from 134,881 HIV-1-infected patients who visited the Third People's Hospital of Shenzhen (Guangdong, China) between January 2011 and June 2022. Only patients with available data on CD4 counts, HIV-RNA and HCMV-DNA copy numbers, and hospitalized stage 2/3 patients with detailed clinical assessments of EODs were included in this study. The chi-square test and Cox regression model were used to examine the association between HCMV-DNA detection and EOD incidence. Longitudinal analysis was performed to examine the effect of anti-HCMV treatment on the incidence of lung and cardiovascular EODs. RESULTS HCMV-DNA had been tested in the blood and urine of 98.6% and 31.8% of the HIV-1-infected patients, respectively. An increased percentage of HCMV was detected in urine (> 2.4-fold) than in blood at different HIV-1 infection stages. In stage 2/3 patients (n = 454), a higher incidence of EODs was observed in those who tested positive for HCMV-DNA in urine (P < 0.0001) than in those who tested positive for HCMV-DNA in blood (P = 0.0977). Using a model for incidence of EODs, we found that HCMV-DNA detection in urine was associated with an increased incidence of lung EOD; the adjusted hazard ratio (HR) was 1.939 (95% confidence interval [CI]: 1.326-2.761, P = 0.0003) for the HCMVurine+ subgroup and 0.933 (95% CI: 0.523-1.623, P = 0.8605) for the HCMVurine- subgroup. A significant HR was also observed for cardiovascular EOD, which was 0.696 (95% CI: 0.492-0.953, P = 0.0302) for the HCMVurine+ group and 1.56 (95% CI: 0.766-3.074, P = 0.2033) for the HCMVurine- group. Longitudinal analysis showed that treatment for HCMV reduced the incidence rates of lung and cardiovascular EODs in the stage 2/3 patients. CONCLUSIONS The presence of HCMV in urine is associated with the early prognosis of EODs in patients with stage 2/3 HIV-1 infection and its detection should be implemented as a routine test.
Collapse
Affiliation(s)
- Fang Zhao
- Department of Infectious Diseases, National Clinical Center for Infectious Diseases, Third People's Hospital of Shenzhen (Second Affiliated Hospital of Southern University of Science and Technology), Shenzhen, Guangdong, PR China
| | - Tsz Yan Fung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region (HKSAR), PR China
| | - Zhiwei Chen
- AIDS Institute, LKS Faculty of Medicine, University of Hong Kong, Pokfulam, HKSAR, PR China; Department of Microbiology, LKS Faculty of Medicine, University of Hong Kong, Pokfulam, HKSAR, PR China.
| | - Hui Wang
- Department of Infectious Diseases, National Clinical Center for Infectious Diseases, Third People's Hospital of Shenzhen (Second Affiliated Hospital of Southern University of Science and Technology), Shenzhen, Guangdong, PR China.
| | - Allen Ka Loon Cheung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region (HKSAR), PR China.
| |
Collapse
|
22
|
Kuan MI, Caruso LB, Zavala AG, Rana PSJB, O'Dowd JM, Tempera I, Fortunato EA. Human Cytomegalovirus Utilizes Multiple Viral Proteins to Regulate the Basement Membrane Protein Nidogen 1. J Virol 2022; 96:e0133622. [PMID: 36218358 PMCID: PMC9599421 DOI: 10.1128/jvi.01336-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 11/20/2022] Open
Abstract
Nidogen 1 (NID1) is an important basement membrane protein secreted by many cell types. We previously found that human cytomegalovirus (HCMV) infection rapidly induced chromosome 1 breaks and that the basement membrane protein NID1, encoded near the 1q42 break site, was downregulated. We have now determined that the specific breaks in and of themselves did not regulate NID1, rather interactions between several viral proteins and the cellular machinery and DNA regulated NID1. We screened a battery of viral proteins present by 24 hours postinfection (hpi) when regulation was induced, including components of the incoming virion and immediate early (IE) proteins. Adenovirus (Ad) delivery of the tegument proteins pp71 and UL35 and the IE protein IE1 influenced steady-state (ss) NID1 levels. IE1's mechanism of regulation was unclear, while UL35 influenced proteasomal regulation of ss NID1. Real-time quantitative PCR (RT-qPCR) experiments determined that pp71 downregulated NID1 transcription. Surprisingly, WF28-71, a fibroblast clone that expresses minute quantities of pp71, suppressed NID1 transcription as efficiently as HCMV infection, resulting in the near absence of ss NID1. Sequence analysis of the region surrounding the 1q42 break sites and NID1 promoter revealed CCCTC-binding factor (CTCF) binding sites. Chromatin immunoprecipitation experiments determined that pp71 and CTCF were both bound at these two sites during HCMV infection. Expression of pp71 alone replicated this binding. Binding was observed as early as 1 hpi, and colocalization of pp71 and CTCF occurred as quickly as 15 min postinfection (pi) in infected cell nuclei. In fibroblasts where CTCF was knocked down, Adpp71 infection did not decrease NID1 transcription nor ss NID1 protein levels. Our results emphasize another aspect of pp71 activity during infection and identify this viral protein as a key contributor to HCMV's efforts to eliminate NID1. Further, we show, for the first time, direct interaction between pp71 and the cellular genome. IMPORTANCE We have found that human cytomegalovirus (HCMV) utilizes multiple viral proteins in multiple pathways to regulate a ubiquitous cellular basement membrane protein, nidogen-1 (NID1). The extent of the resources and the redundant methods that the virus has evolved to affect this control strongly suggest that its removal provides a life cycle advantage to HCMV. Our discoveries that one of the proteins that HCMV uses to control NID1, pp71, binds directly to the cellular DNA and can exert control when present in vanishingly small quantities may have broad implications in a wide range of infection scenarios. Dysregulation of NID1 in an immunocompetent host is not known to manifest complications during infection; however, in the naive immune system of a developing fetus, disruption of this developmentally critical protein could initiate catastrophic HCMV-induced birth defects.
Collapse
Affiliation(s)
- Man I Kuan
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, Idaho, USA
| | | | - Anamaria G. Zavala
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, Idaho, USA
| | - Pranav S. J. B. Rana
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, Idaho, USA
| | - John M. O'Dowd
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, Idaho, USA
| | - Italo Tempera
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Elizabeth A. Fortunato
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, Idaho, USA
| |
Collapse
|
23
|
Perera MR, Sinclair JH. The Human Cytomegalovirus β2.7 Long Non-Coding RNA Prevents Induction of Reactive Oxygen Species to Maintain Viral Gene Silencing during Latency. Int J Mol Sci 2022; 23:ijms231911017. [PMID: 36232315 PMCID: PMC9569889 DOI: 10.3390/ijms231911017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/06/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a significant source of disease for the immunosuppressed and immunonaive. The treatment of HCMV is made more problematic by viral latency, a lifecycle stage in which the virus reduces its own gene expression and produces no infectious virus. The most highly expressed viral gene during HCMV latency is the viral β2.7 long non-coding RNA. Although we have recently shown that the β2.7 lncRNA lowers levels of reactive oxygen species (ROS) during infection in monocytes, how this impacts latency is unclear. We now show that β2.7 is important for establishing and maintaining HCMV latency by aiding the suppression of viral lytic gene expression and that this is directly related to its ability to quench reactive oxygen species (ROS). Consistent with this, we also find that exogenous inducers of ROS cause reactivation of latent HCMV. These effects can be compensated by treatment with an antioxidant to lower ROS levels. Finally, we show that ROS-mediated reactivation is independent of myeloid differentiation, but instead relies on NF-κB activation. Altogether, these results reveal a novel factor that is central to the complex process that underpins HCMV latency. These findings may be of particular relevance in the transplant setting, in which transplanted tissue/organs are subject to very high ROS levels, and HCMV reactivation poses a significant threat.
Collapse
|
24
|
Li N, Wang Z, Wang B, Wang J, Xu R, Yang T, Huang S, Wang H, Yu Q. Identification and Characterization of Long Non-coding RNA in Tomato Roots Under Salt Stress. FRONTIERS IN PLANT SCIENCE 2022; 13:834027. [PMID: 35865296 PMCID: PMC9295719 DOI: 10.3389/fpls.2022.834027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 06/14/2022] [Indexed: 06/15/2023]
Abstract
As one of the most important vegetable crops in the world, the production of tomatoes was restricted by salt stress. Therefore, it is of great interest to analyze the salt stress tolerance genes. As the non-coding RNAs (ncRNAs) with a length of more than 200 nucleotides, long non-coding RNAs (lncRNAs) lack the ability of protein-coding, but they can play crucial roles in plant development and response to abiotic stresses by regulating gene expression. Nevertheless, there are few studies on the roles of salt-induced lncRNAs in tomatoes. Therefore, we selected wild tomato Solanum pennellii (S. pennellii) and cultivated tomato M82 to be materials. By high-throughput sequencing, 1,044 putative lncRNAs were identified here. Among them, 154 and 137 lncRNAs were differentially expressed in M82 and S. pennellii, respectively. Through functional analysis of target genes of differentially expressed lncRNAs (DE-lncRNAs), some genes were found to respond positively to salt stress by participating in abscisic acid (ABA) signaling pathway, brassinosteroid (BR) signaling pathway, ethylene (ETH) signaling pathway, and anti-oxidation process. We also construct a salt-induced lncRNA-mRNA co-expression network to dissect the putative mechanisms of high salt tolerance in S. pennellii. We analyze the function of salt-induced lncRNAs in tomato roots at the genome-wide levels for the first time. These results will contribute to understanding the molecular mechanisms of salt tolerance in tomatoes from the perspective of lncRNAs.
Collapse
Affiliation(s)
- Ning Li
- Institute of Horticulture Crops, Xinjiang Academy of Agricultural Sciences, Urumqi, China
- Key Laboratory of Horticulture Crop Genomics and Genetic Improvement in Xinjiang, Urumqi, China
| | - Zhongyu Wang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Baike Wang
- Institute of Horticulture Crops, Xinjiang Academy of Agricultural Sciences, Urumqi, China
- Key Laboratory of Horticulture Crop Genomics and Genetic Improvement in Xinjiang, Urumqi, China
| | - Juan Wang
- Institute of Horticulture Crops, Xinjiang Academy of Agricultural Sciences, Urumqi, China
- Key Laboratory of Horticulture Crop Genomics and Genetic Improvement in Xinjiang, Urumqi, China
| | - Ruiqiang Xu
- Institute of Horticulture Crops, Xinjiang Academy of Agricultural Sciences, Urumqi, China
- Key Laboratory of Horticulture Crop Genomics and Genetic Improvement in Xinjiang, Urumqi, China
| | - Tao Yang
- Institute of Horticulture Crops, Xinjiang Academy of Agricultural Sciences, Urumqi, China
- Key Laboratory of Horticulture Crop Genomics and Genetic Improvement in Xinjiang, Urumqi, China
| | - Shaoyong Huang
- Institute of Horticulture Crops, Xinjiang Academy of Agricultural Sciences, Urumqi, China
- Key Laboratory of Horticulture Crop Genomics and Genetic Improvement in Xinjiang, Urumqi, China
| | - Huan Wang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qinghui Yu
- Institute of Horticulture Crops, Xinjiang Academy of Agricultural Sciences, Urumqi, China
- Key Laboratory of Horticulture Crop Genomics and Genetic Improvement in Xinjiang, Urumqi, China
| |
Collapse
|
25
|
Yang S, Liu X, Wang M, Cao D, Jaijyan DK, Enescu N, Liu J, Wu S, Wang S, Sun W, Xiao L, Gu A, Li Y, Zhou H, Tyagi S, Wu J, Tang Q, Zhu H. Circular RNAs Represent a Novel Class of Human Cytomegalovirus Transcripts. Microbiol Spectr 2022; 10:e0110622. [PMID: 35604147 PMCID: PMC9241847 DOI: 10.1128/spectrum.01106-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 01/03/2023] Open
Abstract
Human cytomegalovirus (HCMV) infects a large portion of the human population globally. Several HCMV-derived noncoding RNAs are involved in the regulation of viral gene expression and the virus life cycle. Here, we reported that circRNAs are a new class of HCMV transcripts. We bioinformatically predict 704 candidate circRNAs encoded by the TB40/E strain and 230 encoded by the HAN strain. We also systematically compare circRNA features, including the breakpoint sequence consensus, strand preference, length distribution, and exon numbers between host genome-encoded circRNAs and viral circRNAs, and showed that the unique characteristics of viral circRNAs are correlated with their genome types. Furthermore, we experimentally confirmed 324 back-splice junctions (BSJs) from three HCMV strains, Towne, TB40/E, and Toledo, and identified 4 representative HCMV circRNAs by RNase R treatment. Interestingly, we also showed that HCMV contains alternative back-splicing circRNAs. We developed a new amplified FISH method that allowed us to visualize circRNAs and quantify the number of circRNA molecules in the infected cells. The competitive endogenous RNA network analysis suggests that HCMV circRNAs play important roles in viral DNA synthesis via circRNA-miRNA-mRNA networks. Our findings highlight that circRNAs are an important component of the HCMV transcriptome that may contribute to viral replication and pathogenesis. IMPORTANCE HCMV infects 40% to 100% of the human population globally and may be a life-threatening pathogen in immunocompromised individuals. CircRNA is a family of unique RNA that is the most newly found and remains unknown in many aspects. Our current studies computationally identified HCMV-encoded circRNAs and confirmed the existence of the HCMV circRNAs in the infected cells. We systematically compared the features between host and different viral circRNAs and found that the unique characteristics of circRNAs were correlated with their genome types. We also first reported that HCMV contained alternative back-splicing circRNAs. More importantly, we developed a new amplified FISH method which allowed us for the first time not only to visualize circRNAs but also to quantify the number of circRNA molecules in the infected cells. This work describes a novel component of HCMV transcriptome bringing a new understanding of HCMV biology and disease.
Collapse
Affiliation(s)
| | | | - Mei Wang
- Jinan University, Guangzhou, Guangdong, China
| | - Di Cao
- Jinan University, Guangzhou, Guangdong, China
| | - Dabbu Kumar Jaijyan
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Nicole Enescu
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Jian Liu
- School of Biological Sciences and Biotechnology, Minnan Normal University, Zhangzhou, Fujian, China
| | - Songbin Wu
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Sashuang Wang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Wuping Sun
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Lizu Xiao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Alison Gu
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Yaolan Li
- Jinan University, Guangzhou, Guangdong, China
| | - Hong Zhou
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Sanjay Tyagi
- Public Health Research Institute, New Jersey Medical School, Newark, New Jersey, USA
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Jianguo Wu
- Jinan University, Guangzhou, Guangdong, China
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Hua Zhu
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
26
|
Yu Z, Wang J, Nan F, Shi W, Zhang X, Jiang S, Wang B. Human Cytomegalovirus Induced Aberrant Expression of Non-coding RNAs. Front Microbiol 2022; 13:918213. [PMID: 35770158 PMCID: PMC9234646 DOI: 10.3389/fmicb.2022.918213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a β-herpesvirus whose genome consists of double stranded linear DNA. HCMV genome can generate non-coding RNAs (ncRNAs) through transcription in its host cells. Besides that, HCMV infection also changes the ncRNAs expression profile of the host cells. ncRNAs play a key role in maintaining the normal physiological activity of cells, and the disorder of ncRNAs expression has numerous adverse effects on cells. However, until now, the relationship between ncRNAs and HCMV-induced adverse effects are not summarized in detail. This review aims to give a systematic summary of the role of HCMV infection in ncRNAs expression while providing insights into the molecular mechanism of unnormal cellular events caused by ncRNAs disorder. ncRNAs disorder induced by HCMV infection is highly associated with cell proliferation, apoptosis, tumorigenesis, and immune regulation, as well as the development of cardiovascular diseases, and the potential role of biomarker. We summarize the studies on HCMV associated ncRNAs disorder and suggest innovative strategies for eliminating the adverse effects caused by HCMV infection.
Collapse
Affiliation(s)
- Zhongjie Yu
- Department of Special Medicine, School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Jing Wang
- Oral Research Center, Qingdao Municipal Hospital, Qingdao, China
| | - Fulong Nan
- Department of Special Medicine, School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Wenyi Shi
- Oral Research Center, Qingdao Municipal Hospital, Qingdao, China
| | - Xianjuan Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Shasha Jiang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Bin Wang
- Department of Special Medicine, School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
- *Correspondence: Bin Wang,
| |
Collapse
|
27
|
Nehme Z, Pasquereau S, Haidar Ahmad S, El Baba R, Herbein G. Polyploid giant cancer cells, EZH2 and Myc upregulation in mammary epithelial cells infected with high-risk human cytomegalovirus. EBioMedicine 2022; 80:104056. [PMID: 35596973 PMCID: PMC9121245 DOI: 10.1016/j.ebiom.2022.104056] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 02/08/2023] Open
Abstract
Background Human cytomegalovirus (HCMV) infection has been actively implicated in complex neoplastic processes. Beyond oncomodulation, the molecular mechanisms that might underlie HCMV-induced oncogenesis are being extensively studied. Polycomb repressive complex 2 (PRC2) proteins, in particular enhancer of zeste homolog 2 (EZH2) are associated with cancer progression. Nevertheless, little is known about EZH2 activation in the context of HCMV infection and breast oncogenesis. Methods Herein, we identified EZH2 as a downstream target for HCMV-induced Myc upregulation upon acute and chronic infection with high-risk strains using a human mammary epithelial model. Findings We detected polyploidy and CMV-transformed HMECs (CTH) cells harboring HCMV and dynamically undergoing the giant cells cycle. Acquisition of embryonic stemness markers positively correlated with EZH2 and Myc expression. EZH2 inhibitors curtail sustained CTH cells’ malignant phenotype. Besides harboring polyploid giant cancer cells (PGCCs), tumorigenic breast biopsies were characterized by an enhanced EZH2 and Myc expression, with a strong positive correlation between EZH2 and Myc expression, and between PGCC count and EZH2/Myc expression in the presence of HCMV. Further, we isolated two HCMV strains from EZH2HighMycHigh basal-like tumors which replicate in MRC5 cells and transform HMECs toward CTH cells after acute infection. Interpretation Our data establish a potential link between HCMV-induced Myc activation, the subsequent EZH2 upregulation, and polyploidy induction. These data support the proposed tumorigenesis properties of EZH2/Myc, and allow the isolation of two oncogenic HCMV strains from EZH2HighMycHigh basal breast tumors while identifying EZH2 as a potential therapeutic target in the management of breast cancer, particularly upon HCMV infection. Funding This work was supported by grants from the University of Franche-Comté (UFC) (CR3300), the Région Franche-Comté (2021-Y-08292 and 2021-Y-08290) and the Ligue contre le Cancer (CR3304) to Georges Herbein. Zeina Nehme is a recipient of a doctoral scholarship from the municipality of Habbouch. Sandy Haidar Ahmad is recipient of a doctoral scholarship from Lebanese municipality. Ranim El Baba is a recipient of a doctoral scholarship from Hariri foundation for sustainable human development.
Collapse
Affiliation(s)
- Zeina Nehme
- Department Pathogens and Inflammation-EPILAB, EA4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), 16 route de Gray, Besançon F-25030, France
| | - Sébastien Pasquereau
- Department Pathogens and Inflammation-EPILAB, EA4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), 16 route de Gray, Besançon F-25030, France
| | - Sandy Haidar Ahmad
- Department Pathogens and Inflammation-EPILAB, EA4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), 16 route de Gray, Besançon F-25030, France
| | - Ranim El Baba
- Department Pathogens and Inflammation-EPILAB, EA4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), 16 route de Gray, Besançon F-25030, France
| | - Georges Herbein
- Department Pathogens and Inflammation-EPILAB, EA4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), 16 route de Gray, Besançon F-25030, France; Department of Virology, CHU Besançon, Besançon, France.
| |
Collapse
|
28
|
Huang Y, Guo X, Zhang J, Li J, Xu M, Wang Q, Liu Z, Ma Y, Qi Y, Ruan Q. Human cytomegalovirus RNA2.7 inhibits RNA polymerase II (Pol II) Serine-2 phosphorylation by reducing the interaction between Pol II and phosphorylated cyclin-dependent kinase 9 (pCDK9). Virol Sin 2022; 37:358-369. [PMID: 35537980 PMCID: PMC9243627 DOI: 10.1016/j.virs.2022.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 02/24/2022] [Indexed: 11/18/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous pathogen belongs to betaherpesvirus subfamily. RNA2.7 is a highly conserved long non-coding RNA accounting for more than 20% of total viral transcripts. In our study, functions of HCMV RNA2.7 were investigated by comparison of host cellular transcriptomes between cells infected with HCMV clinical strain and RNA2.7 deleted mutant. It was demonstrated that RNA polymerase II (Pol II)-dependent host gene transcriptions were significantly activated when RNA2.7 was removed during infection. A 145 nt-in-length motif within RNA2.7 was identified to inhibit the phosphorylation of Pol II Serine-2 (Pol II S2) by reducing the interaction between Pol II and phosphorylated cyclin-dependent kinase 9 (pCDK9). Due to the loss of Pol II S2 phosphorylation, cellular DNA pre-replication complex (pre-RC) factors, including Cdt1 and Cdc6, were significantly decreased, which prevented more cells from entering into S phase and facilitated viral DNA replication. Our results provide new insights of HCMV RNA2.7 functions in regulation of host cellular transcription. HCMV RNA2.7 inhibits the phosphorylation of Pol II Serine-2. RNA2.7 reduces the interactions between Pol II and pCDK9. RNA2.7 regulates cell cycle by preventing cells from entering into S phase.
Collapse
Affiliation(s)
- Yujing Huang
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xin Guo
- Department of Pediatrics, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110033, China
| | - Jing Zhang
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Jianming Li
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Mingyi Xu
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Qing Wang
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Zhongyang Liu
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yanping Ma
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Ying Qi
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Qiang Ruan
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
29
|
A Viral Long Non-Coding RNA Protects against Cell Death during Human Cytomegalovirus Infection of CD14+ Monocytes. Viruses 2022; 14:v14020246. [PMID: 35215840 PMCID: PMC8874509 DOI: 10.3390/v14020246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 02/07/2023] Open
Abstract
Long non-coding RNA β2.7 is the most highly transcribed viral gene during latent human cytomegalovirus (HCMV) infection. However, as yet, no function has ever been ascribed to β2.7 during HCMV latency. Here we show that β2.7 protects against apoptosis induced by high levels of reactive oxygen species (ROS) in infected monocytes, which routinely support latent HCMV infection. Monocytes infected with a wild-type (WT) virus, but not virus deleted for the β2.7 gene (Δβ2.7), are protected against mitochondrial stress and subsequent apoptosis. Protected monocytes display lower levels of ROS and additionally, stress-induced death in the absence of β2.7 can be reversed by an antioxidant which reduces ROS levels. Furthermore, we show that infection with WT but not Δβ2.7 virus results in strong upregulation of a cellular antioxidant enzyme, superoxide dismutase 2 (SOD2) in CD14+ monocytes. These observations identify a role for the β2.7 viral transcript, the most abundantly expressed viral RNA during latency but for which no latency-associated function has ever been ascribed, and demonstrate a novel way in which HCMV protects infected monocytes from pro-death signals to optimise latent carriage.
Collapse
|
30
|
Manska S, Rossetto CC. Identification of cellular proteins associated with human cytomegalovirus (HCMV) DNA replication suggests novel cellular and viral interactions. Virology 2022; 566:26-41. [PMID: 34861458 PMCID: PMC8720285 DOI: 10.1016/j.virol.2021.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/07/2021] [Accepted: 11/16/2021] [Indexed: 01/03/2023]
Abstract
Upon entry of Human cytomegalovirus (HCMV) into the host cell, the viral genome is transported to the nucleus where it serves as a template for transcription and genome replication. Production of new viral genomes is a coordinated effort between viral and cellular proteins. While the core replication proteins are encoded by the virus, additional cellular proteins support the process of genome synthesis. We used accelerated native isolation of proteins on nascent DNA (aniPOND) to study protein dynamics on nascent viral DNA during HCMV infection. Using this method, we identified specific viral and cellular proteins that are associated with nascent viral DNA. These included transcription factors, transcriptional regulators, DNA damage and repair factors, and chromatin remodeling complexes. The association of these identified proteins with viral DNA was confirmed by immunofluorescent imaging, chromatin-immunoprecipitation analyses, and shRNA knockdown experiments. These data provide evidence for the requirement of cellular factors involved in HCMV replication.
Collapse
Affiliation(s)
- Salomé Manska
- University of Nevada, Reno School of Medicine, Department of Microbiology and Immunology, 1664 North Virginia Street/MS320, Reno, NV 89557 USA
| | - Cyprian C. Rossetto
- University of Nevada, Reno School of Medicine, Department of Microbiology and Immunology, 1664 North Virginia Street/MS320, Reno, NV 89557 USA,Correspondence to: Cyprian C. Rossetto, Ph.D.
| |
Collapse
|
31
|
Lau B, Kerr K, Camiolo S, Nightingale K, Gu Q, Antrobus R, Suárez NM, Loney C, Stanton RJ, Weekes MP, Davison AJ. Human Cytomegalovirus RNA2.7 Is Required for Upregulating Multiple Cellular Genes To Promote Cell Motility and Viral Spread Late in Lytic Infection. J Virol 2021; 95:e0069821. [PMID: 34346763 PMCID: PMC8475523 DOI: 10.1128/jvi.00698-21] [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: 04/25/2021] [Accepted: 07/22/2021] [Indexed: 11/20/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are frequently associated with broad modulation of gene expression and thus provide the cell with the ability to synchronize entire metabolic processes. We used transcriptomic approaches to investigate whether the most abundant human cytomegalovirus-encoded lncRNA, RNA2.7, has this characteristic. By comparing cells infected with wild-type virus (WT) to cells infected with RNA2.7 deletion mutants, RNA2.7 was implicated in regulating a large number of cellular genes late in lytic infection. Pathway analysis indicated that >100 of these genes are associated with promoting cell movement, and the 10 most highly regulated of these were validated in further experiments. Morphological analysis and live cell tracking of WT- and RNA2.7 mutant-infected cells indicated that RNA2.7 is involved in promoting the movement and detachment of infected cells late in infection, and plaque assays using sparse cell monolayers indicated that RNA2.7 is also involved in promoting cell-to-cell spread of virus. Consistent with the observation that upregulated mRNAs are relatively A+U-rich, which is a trait associated with transcript instability, and that they are also enriched in motifs associated with mRNA instability, transcriptional inhibition experiments on WT- and RNA2.7 mutant-infected cells showed that four upregulated transcripts lived longer in the presence of RNA2.7. These findings demonstrate that RNA2.7 is required for promoting cell movement and viral spread late in infection and suggest that this may be due to general stabilization of A+U-rich transcripts. IMPORTANCE In addition to messenger RNAs (mRNAs), the human genome encodes a large number of long noncoding RNAs (lncRNAs). Many lncRNAs that have been studied in detail are associated with broad modulation of gene expression and have important biological roles. Human cytomegalovirus, which is a large, clinically important DNA virus, specifies four lncRNAs, one of which (RNA2.7) is expressed at remarkably high levels during lytic infection. Our studies show that RNA2.7 is required for upregulating a large number of human genes, about 100 of which are associated with cell movement, and for promoting the movement of infected cells and the spread of virus from one cell to another. Further bioinformatic and experimental analyses indicated that RNA2.7 may exert these effects by stabilizing mRNAs that are relatively rich in A and U nucleotides. These findings increase our knowledge of how human cytomegalovirus regulates the infected cell to promote its own success.
Collapse
Affiliation(s)
- Betty Lau
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Karen Kerr
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Salvatore Camiolo
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Katie Nightingale
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Quan Gu
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Nicolás M. Suárez
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Colin Loney
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Richard J. Stanton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Michael P. Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Andrew J. Davison
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| |
Collapse
|
32
|
De Novo Polycomb Recruitment: Lessons from Latent Herpesviruses. Viruses 2021; 13:v13081470. [PMID: 34452335 PMCID: PMC8402699 DOI: 10.3390/v13081470] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 12/11/2022] Open
Abstract
The Human Herpesviruses persist in the form of a latent infection in specialized cell types. During latency, the herpesvirus genomes associate with cellular histone proteins and the viral lytic genes assemble into transcriptionally repressive heterochromatin. Although there is divergence in the nature of heterochromatin on latent herpesvirus genomes, in general, the genomes assemble into forms of heterochromatin that can convert to euchromatin to permit gene expression and therefore reactivation. This reversible form of heterochromatin is known as facultative heterochromatin and is most commonly characterized by polycomb silencing. Polycomb silencing is prevalent on the cellular genome and plays a role in developmentally regulated and imprinted genes, as well as X chromosome inactivation. As herpesviruses initially enter the cell in an un-chromatinized state, they provide an optimal system to study how de novo facultative heterochromatin is targeted to regions of DNA and how it contributes to silencing. Here, we describe how polycomb-mediated silencing potentially assembles onto herpesvirus genomes, synergizing what is known about herpesvirus latency with facultative heterochromatin targeting to the cellular genome. A greater understanding of polycomb silencing of herpesviruses will inform on the mechanism of persistence and reactivation of these pathogenic human viruses and provide clues regarding how de novo facultative heterochromatin forms on the cellular genome.
Collapse
|
33
|
Viral Manipulation of the Host Epigenome as a Driver of Virus-Induced Oncogenesis. Microorganisms 2021; 9:microorganisms9061179. [PMID: 34070716 PMCID: PMC8227491 DOI: 10.3390/microorganisms9061179] [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: 04/28/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Tumorigenesis due to viral infection accounts for a high fraction of the total global cancer burden (15–20%) of all human cancers. A comprehensive understanding of the mechanisms by which viral infection leads to tumor development is extremely important. One of the main mechanisms by which viruses induce host cell proliferation programs is through controlling the host’s epigenetic machinery. In this review, we dissect the epigenetic pathways through which oncogenic viruses can integrate their genome into host cell chromosomes and lead to tumor progression. In addition, we highlight the potential use of drugs based on histone modifiers in reducing the global impact of cancer development due to viral infection.
Collapse
|
34
|
Epigenetic reprogramming of host and viral genes by Human Cytomegalovirus infection in Kasumi-3 myeloid progenitor cells at early times post-infection. J Virol 2021; 95:JVI.00183-21. [PMID: 33731453 PMCID: PMC10021080 DOI: 10.1128/jvi.00183-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HCMV establishes latency in myeloid cells. Using the Kasumi-3 latency model, we previously showed that lytic gene expression is activated prior to establishment of latency in these cells. The early events in infection may have a critical role in shaping establishment of latency. Here, we have used an integrative multi-omics approach to investigate dynamic changes in host and HCMV gene expression and epigenomes at early times post infection. Our results show dynamic changes in viral gene expression and viral chromatin. Analyses of Pol II, H3K27Ac and H3K27me3 occupancy of the viral genome showed that 1) Pol II occupancy was highest at the MIEP at 4 hours post infection. However, it was observed throughout the genome; 2) At 24 hours, H3K27Ac was localized to the major immediate early promoter/enhancer and to a possible second enhancer in the origin of replication OriLyt; 3) viral chromatin was broadly accessible at 24 hpi. In addition, although HCMV infection activated expression of some host genes, we observed an overall loss of de novo transcription. This was associated with loss of promoter-proximal Pol II and H3K27Ac, but not with changes in chromatin accessibility or a switch in modification of H3K27.Importance.HCMV is an important human pathogen in immunocompromised hosts and developing fetuses. Current anti-viral therapies are limited by toxicity and emergence of resistant strains. Our studies highlight emerging concepts that challenge current paradigms of regulation of HCMV gene expression in myeloid cells. In addition, our studies show that HCMV has a profound effect on de novo transcription and the cellular epigenome. These results may have implications for mechanisms of viral pathogenesis.
Collapse
|
35
|
The Zinc Finger Antiviral Protein ZAP Restricts Human Cytomegalovirus and Selectively Binds and Destabilizes Viral UL4/ UL5 Transcripts. mBio 2021; 12:mBio.02683-20. [PMID: 33947766 PMCID: PMC8263000 DOI: 10.1128/mbio.02683-20] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Interferon-stimulated gene products (ISGs) play a crucial role in early infection control. The ISG zinc finger CCCH-type antiviral protein 1 (ZAP/ZC3HAV1) antagonizes several RNA viruses by binding to CG-rich RNA sequences, whereas its effect on DNA viruses is less well understood. Here, we decipher the role of ZAP in the context of human cytomegalovirus (HCMV) infection, a β-herpesvirus that is associated with high morbidity in immunosuppressed individuals and newborns. We show that expression of the two major isoforms of ZAP, ZAP-S and ZAP-L, is induced during HCMV infection and that both negatively affect HCMV replication. Transcriptome and proteome analyses demonstrated that the expression of ZAP results in reduced viral mRNA and protein levels and decelerates the progression of HCMV infection. Metabolic RNA labeling combined with high-throughput sequencing (SLAM-seq) revealed that most of the gene expression changes late in infection result from the general attenuation of HCMV. Furthermore, at early stages of infection, ZAP restricts HCMV by destabilizing a distinct subset of viral mRNAs, particularly those from the previously uncharacterized UL4-UL6 HCMV gene locus. Through enhanced cross-linking immunoprecipitation and sequencing analysis (eCLIP-seq), we identified the transcripts expressed from this HCMV locus as the direct targets of ZAP. Moreover, our data show that ZAP preferentially recognizes not only CG, but also other cytosine-rich sequences, thereby expanding its target specificity. In summary, this report is the first to reveal direct targets of ZAP during HCMV infection, which strongly indicates that transcripts from the UL4-UL6 locus may play an important role for HCMV replication.IMPORTANCE Viral infections have a large impact on society, leading to major human and economic losses and even global instability. So far, many viral infections, including human cytomegalovirus (HCMV) infection, are treated with a small repertoire of drugs, often accompanied by the occurrence of resistant mutants. There is no licensed HCMV vaccine in sight to protect those most at risk, particularly immunocompromised individuals or pregnant women who might otherwise transmit the virus to the fetus. Thus, the identification of novel intervention strategies is urgently required. In this study, we show that ZAP decelerates the viral gene expression cascade, presumably by selectively handpicking a distinct set of viral transcripts for degradation. Our study illustrates the potent role of ZAP as an HCMV restriction factor and sheds light on a possible role for UL4 and/or UL5 early during infection, paving a new avenue for the exploration of potential targets for novel therapies.
Collapse
|
36
|
Perera MR, Wills MR, Sinclair JH. HCMV Antivirals and Strategies to Target the Latent Reservoir. Viruses 2021; 13:817. [PMID: 34062863 PMCID: PMC8147263 DOI: 10.3390/v13050817] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/11/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous human herpesvirus. In healthy people, primary infection is generally asymptomatic, and the virus can go on to establish lifelong latency in cells of the myeloid lineage. However, HCMV often causes severe disease in the immunosuppressed: transplant recipients and people living with AIDS, and also in the immunonaive foetus. At present, there are several antiviral drugs licensed to control HCMV disease. However, these are all faced with problems of poor bioavailability, toxicity and rapidly emerging viral resistance. Furthermore, none of them are capable of fully clearing the virus from the host, as they do not target latent infection. Consequently, reactivation from latency is a significant source of disease, and there remains an unmet need for treatments that also target latent infection. This review briefly summarises the most common HCMV antivirals used in clinic at present and discusses current research into targeting the latent HCMV reservoir.
Collapse
Affiliation(s)
| | | | - John H. Sinclair
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, UK; (M.R.P.); (M.R.W.)
| |
Collapse
|
37
|
Elder EG, Krishna BA, Poole E, Perera M, Sinclair J. Regulation of host and viral promoters during human cytomegalovirus latency via US28 and CTCF. J Gen Virol 2021; 102:001609. [PMID: 34042564 PMCID: PMC8295918 DOI: 10.1099/jgv.0.001609] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Viral latency is an active process during which the host cell environment is optimized for latent carriage and reactivation. This requires control of both viral and host gene promoters and enhancers often at the level of chromatin, and several viruses co-opt the chromatin organiser CTCF to control gene expression during latency. While CTCF has a role in the latencies of alpha- and gamma-herpesviruses, it was not known whether CTCF played a role in the latency of the beta-herpesvirus human cytomegalovirus (HCMV). Here, we show that HCMV latency is associated with increased CTCF expression and CTCF binding to the viral major lytic promoter, the major immediate early promoter (MIEP). This increase in CTCF binding is dependent on the virally encoded G protein coupled receptor, US28, and contributes to suppression of MIEP-driven transcription, a hallmark of latency. Furthermore, we show that latency-associated upregulation of CTCF represses expression of the neutrophil chemoattractants S100A8 and S100A9 which we have previously shown are downregulated during HCMV latency. As with downregulation of the MIEP, CTCF binding to the enhancer region of S100A8/A9 drives their suppression, again in a US28-dependent manner. Taken together, we identify CTCF upregulation as an important mechanism for optimizing latent carriage of HCMV at both the levels of viral and cellular gene expression.
Collapse
Affiliation(s)
- Elizabeth G. Elder
- Department of Medicine, University of Cambridge, Cambridge, UK
- Present address: Public Health Agency of Sweden, Solna, Sweden
| | | | - Emma Poole
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Marianne Perera
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - John Sinclair
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
38
|
Haidar Ahmad S, Al Moussawi F, El Baba R, Nehme Z, Pasquereau S, Kumar A, Molimard C, Monnien F, Algros MP, Karaky R, Stamminger T, Diab Assaf M, Herbein G. Identification of UL69 Gene and Protein in Cytomegalovirus-Transformed Human Mammary Epithelial Cells. Front Oncol 2021; 11:627866. [PMID: 33937031 PMCID: PMC8085531 DOI: 10.3389/fonc.2021.627866] [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: 11/10/2020] [Accepted: 03/26/2021] [Indexed: 12/15/2022] Open
Abstract
A growing body of evidence addressing the involvement of human cytomegalovirus (HCMV) in malignancies had directed attention to the oncomodulation paradigm. HCMV-DB infected human mammary epithelial cells (HMECs) in culture showed the emergence of clusters of rapidly proliferating, spheroid-shaped transformed cells named CTH (CMV-Transformed HMECs) cells. CTH cells assessment suggests a direct contribution of HCMV to oncogenesis, from key latent and lytic genes activating oncogenic pathways to fueling tumor evolution. We hypothesized that the presence of HCMV genome in CTH cells is of pivotal importance for determining its oncogenic potential. We previously reported the detection of a long non-coding (lnc) RNA4.9 gene in CTH cells. Therefore, we assessed here the presence of UL69 gene, located nearby and downstream of the lncRNA4.9 gene, in CTH cells. The HCMV UL69 gene in CTH cells was detected using polymerase chain reaction (PCR) and sequencing of UL69 gene was performed using Sanger method. The corresponding amino acid sequence was then blasted against the UL69 sequence derived from HCMV-DB genome using NCBI Protein BLAST tool. A 99% identity was present between the nucleotide sequence present in CTH cells and HCMV-DB genome. UL69 transcript was detected in RNA extracts of CTH cells, using a reverse transcription polymerase chain reaction (RT-PCR) assay, and pUL69 protein was identified in CTH lysates using western blotting. Ganciclovir-treated CTH cells showed a decrease in UL69 gene detection and cellular proliferation. In CTH cells, the knockdown of UL69 with siRNA was assessed by RT-qPCR and western blot to reveal the impact of pUL69 on HCMV replication and CTH cell proliferation. Finally, UL69 gene was detected in breast cancer biopsies. Our results indicate a close link between the UL69 gene detected in the HCMV-DB isolate used to infect HMECs, and the UL69 gene present in transformed CTH cells and tumor biopsies, further highlighting a direct role for HCMV in breast tumor development.
Collapse
Affiliation(s)
- Sandy Haidar Ahmad
- Department Pathogens & Inflammation-EPILAB EA4266, University of Bourgogne France-Comté, Besançon, France.,Molecular Cancer and Pharmaceutical Biology Laboratory, Lebanese University, Beyrouth, Lebanon
| | - Fatima Al Moussawi
- Department Pathogens & Inflammation-EPILAB EA4266, University of Bourgogne France-Comté, Besançon, France.,Molecular Cancer and Pharmaceutical Biology Laboratory, Lebanese University, Beyrouth, Lebanon
| | - Ranim El Baba
- Department Pathogens & Inflammation-EPILAB EA4266, University of Bourgogne France-Comté, Besançon, France.,Molecular Cancer and Pharmaceutical Biology Laboratory, Lebanese University, Beyrouth, Lebanon
| | - Zeina Nehme
- Department Pathogens & Inflammation-EPILAB EA4266, University of Bourgogne France-Comté, Besançon, France.,Molecular Cancer and Pharmaceutical Biology Laboratory, Lebanese University, Beyrouth, Lebanon
| | - Sébastien Pasquereau
- Department Pathogens & Inflammation-EPILAB EA4266, University of Bourgogne France-Comté, Besançon, France
| | - Amit Kumar
- Department Pathogens & Inflammation-EPILAB EA4266, University of Bourgogne France-Comté, Besançon, France
| | - Chloé Molimard
- Department of Pathology, CHRU Besançon, Besançon, France
| | - Franck Monnien
- Department of Pathology, CHRU Besançon, Besançon, France
| | | | - Racha Karaky
- Molecular Cancer and Pharmaceutical Biology Laboratory, Lebanese University, Beyrouth, Lebanon
| | | | - Mona Diab Assaf
- Molecular Cancer and Pharmaceutical Biology Laboratory, Lebanese University, Beyrouth, Lebanon
| | - Georges Herbein
- Department Pathogens & Inflammation-EPILAB EA4266, University of Bourgogne France-Comté, Besançon, France.,Department of Virology, CHRU Besancon, Besancon, France
| |
Collapse
|
39
|
Jackson SE, Chen KC, Groves IJ, Sedikides GX, Gandhi A, Houldcroft CJ, Poole EL, Montanuy I, Mason GM, Okecha G, Reeves MB, Sinclair JH, Wills MR. Latent Cytomegalovirus-Driven Recruitment of Activated CD4+ T Cells Promotes Virus Reactivation. Front Immunol 2021; 12:657945. [PMID: 33912186 PMCID: PMC8072157 DOI: 10.3389/fimmu.2021.657945] [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: 01/24/2021] [Accepted: 03/19/2021] [Indexed: 12/15/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection is not cleared by the initial immune response but persists for the lifetime of the host, in part due to its ability to establish a latent infection in cells of the myeloid lineage. HCMV has been shown to manipulate the secretion of cellular proteins during both lytic and latent infection; with changes caused by latent infection mainly investigated in CD34+ progenitor cells. Whilst CD34+ cells are generally bone marrow resident, their derivative CD14+ monocytes migrate to the periphery where they briefly circulate until extravasation into tissue sites. We have analyzed the effect of HCMV latent infection on the secretome of CD14+ monocytes, identifying an upregulation of both CCL8 and CXCL10 chemokines in the CD14+ latency-associated secretome. Unlike CD34+ cells, the CD14+ latency-associated secretome did not induce migration of resting immune cell subsets but did induce migration of activated NK and T cells expressing CXCR3 in a CXCL10 dependent manner. As reported in CD34+ latent infection, the CD14+ latency-associated secretome also suppressed the anti-viral activity of stimulated CD4+ T cells. Surprisingly, however, co-culture of activated autologous CD4+ T cells with latently infected monocytes resulted in reactivation of HCMV at levels comparable to those observed using M-CSF and IL-1β cytokines. We propose that these events represent a potential strategy to enable HCMV reactivation and local dissemination of the virus at peripheral tissue sites.
Collapse
Affiliation(s)
- Sarah E Jackson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Kevin C Chen
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Ian J Groves
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - George X Sedikides
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Amar Gandhi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Charlotte J Houldcroft
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Emma L Poole
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Inmaculada Montanuy
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Gavin M Mason
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Georgina Okecha
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Matthew B Reeves
- Institute of Immunity & Transplantation, University College London (UCL), London, United Kingdom
| | - John H Sinclair
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Mark R Wills
- Cambridge Institute of Therapeutic Immunology and Infectious Disease and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| |
Collapse
|
40
|
Hull R, Mbita Z, Dlamini Z. Long non-coding RNAs (LncRNAs), viral oncogenomics, and aberrant splicing events: therapeutics implications. Am J Cancer Res 2021; 11:866-883. [PMID: 33791160 PMCID: PMC7994164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/21/2020] [Indexed: 06/12/2023] Open
Abstract
It has been estimated that worldwide up to 10% of all human cancers are the result of viral infection, with 7.2% of all cancers in the developed world have a viral aetiology. In contrast, 22.9% of infections in the developing world are the result of viral infections. This number increases to 30% in Sub-Saharan Africa. The ability of viral infections to induce the transformation of normal cells into cancerous cells is well documented. These viruses are mainly Hepatitis B and C viruses, Epstein Barr virus, Human papillomavirus and Human Cytomegalovirus. They can induce the transformation of normal cells into cancer cells and this may be the underlying cause of carcinogenesis in many different types of cancer. These include liver cancer, lymphoma, nasopharyngeal cancer, cervical cancer, gastric cancer and even glioblastoma. Long non-coding RNAs (LncRNAs) can function by regulating the expression of their target genes by controlling the stability of the target mRNAs or by blocking translation of the target mRNA. They can control transcription by regulating the recruitment of transcription factors or chromatin modification complexes. Finally, lncRNAs can control the phosphorylation, acetylation, and ubiquitination of proteins at the post-translation level. Thus, altering protein localisation, function, folding, stability and ultimately expression. In addition to these functions, lncRNA also regulate alternate pre-mRNA splicing in ways that contribute to the formation of tumours. This mainly involves the interaction of lncRNAs with splicing factors, which alters their activity and function. The ability of lncRNAs to regulate the stability, expression and function of tumour suppressor proteins is important in the development and progression of cancers. LncRNAs also regulate viral replication and latency, leading to carcinogenesis. These factors all make lncRNAs ideal targets for the development of biomarker arrays that can be based on secreted lncRNAs leading to the development of affordable non-invasive biomarker tests for the stage specific diagnosis of tumours. These lncRNAs can also serve as targets for the development of new anticancer drug treatments.
Collapse
Affiliation(s)
- Rodney Hull
- SA-MRC/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, University of Pretoria Hatfield0028, South Africa
| | - Zukile Mbita
- Department of Biochemistry, Microbiology and Biochemistry, University of LimpopoSovenga 0727, South Africa
| | - Zodwa Dlamini
- SA-MRC/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, University of Pretoria Hatfield0028, South Africa
| |
Collapse
|
41
|
Diggins NL, Skalsky RL, Hancock MH. Regulation of Latency and Reactivation by Human Cytomegalovirus miRNAs. Pathogens 2021; 10:pathogens10020200. [PMID: 33668486 PMCID: PMC7918750 DOI: 10.3390/pathogens10020200] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022] Open
Abstract
Human cytomegalovirus (HCMV) encodes 22 mature microRNAs (miRNAs), which regulate a myriad of cellular processes, including vesicular trafficking, cell cycle progression, apoptosis, and immune evasion, as well as viral gene expression. Recent evidence points to a critical role for HCMV miRNAs in mediating latency in CD34+ hematopoietic progenitor cells through modulation of cellular signaling pathways, including attenuation of TGFβ and EGFR signaling. Moreover, HCMV miRNAs can act in concert with, or in opposition to, viral proteins in regulating host cell functions. Here, we comprehensively review the studies of HCMV miRNAs in the context of latency and highlight the novel processes that are manipulated by the virus using these small non-coding RNAs.
Collapse
|
42
|
Abstract
Herpesviruses infect virtually all humans and establish lifelong latency and reactivate to infect other humans. Latency requires multiple functions: maintaining the herpesvirus genome in the nuclei of cells; partitioning the viral genome to daughter cells in dividing cells; avoiding recognition by the immune system by limiting protein expression; producing noncoding viral RNAs (including microRNAs) to suppress lytic gene expression or regulate cellular protein expression that could otherwise eliminate virus-infected cells; modulating the epigenetic state of the viral genome to regulate viral gene expression; and reactivating to infect other hosts. Licensed antivirals inhibit virus replication, but do not affect latency. Understanding of the mechanisms of latency is leading to novel approaches to destroy latently infected cells or inhibit reactivation from latency.
Collapse
|
43
|
CD34 + Hematopoietic Progenitor Cell Subsets Exhibit Differential Ability To Maintain Human Cytomegalovirus Latency and Persistence. J Virol 2021; 95:JVI.02105-20. [PMID: 33177198 DOI: 10.1128/jvi.02105-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 11/20/2022] Open
Abstract
In human cytomegalovirus (HCMV)-seropositive patients, CD34+ hematopoietic progenitor cells (HPCs) provide an important source of latent virus that reactivates following cellular differentiation into tissue macrophages. Multiple groups have used primary CD34+ HPCs to investigate mechanisms of viral latency. However, analyses of mechanisms of HCMV latency have been hampered by the genetic variability of CD34+ HPCs from different donors, availability of cells, and low frequency of reactivation. In addition, multiple progenitor cell types express surface CD34, and the frequencies of these populations differ depending on the tissue source of the cells and culture conditions in vitro In this study, we generated CD34+ progenitor cells from two different embryonic stem cell (ESC) lines, WA01 and WA09, to circumvent limitations associated with primary CD34+ HPCs. HCMV infection of CD34+ HPCs derived from either WA01 or WA09 ESCs supported HCMV latency and induced myelosuppression similar to infection of primary CD34+ HPCs. Analysis of HCMV-infected primary or ESC-derived CD34+ HPC subpopulations indicated that HCMV was able to establish latency and reactivate in CD38+ CD90+ and CD38+/low CD90- HPCs but persistently infected CD38- CD90+ cells to produce infectious virus. These results indicate that ESC-derived CD34+ HPCs can be used as a model for HCMV latency and that the virus either latently or persistently infects specific subpopulations of CD34+ cells.IMPORTANCE Human cytomegalovirus infection is associated with severe disease in transplant patients and understanding how latency and reactivation occur in stem cell populations is essential to understand disease. CD34+ hematopoietic progenitor cells (HPCs) are a critical viral reservoir; however, these cells are a heterogeneous pool with donor-to-donor variation in functional, genetic, and phenotypic characteristics. We generated a novel system using embryonic stem cell lines to model HCMV latency and reactivation in HPCs with a consistent cellular background. Our study defined three key stem cell subsets with differentially regulated latent and replicative states, which provide cellular candidates for isolation and treatment of transplant-mediated disease. This work provides a direction toward developing strategies to control the switch between latency and reactivation.
Collapse
|
44
|
Wen Y, Chen H, Luo F, Zhou H, Li Z. Roles of long noncoding RNAs in bacterial infection. Life Sci 2020; 263:118579. [DOI: 10.1016/j.lfs.2020.118579] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/28/2020] [Accepted: 10/03/2020] [Indexed: 02/09/2023]
|
45
|
Regulation of the MIE Locus During HCMV Latency and Reactivation. Pathogens 2020; 9:pathogens9110869. [PMID: 33113934 PMCID: PMC7690695 DOI: 10.3390/pathogens9110869] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous herpesviral pathogen that results in life-long infection. HCMV maintains a latent or quiescent infection in hematopoietic cells, which is broadly defined by transcriptional silencing and the absence of de novo virion production. However, upon cell differentiation coupled with immune dysfunction, the virus can reactivate, which leads to lytic replication in a variety of cell and tissue types. One of the mechanisms controlling the balance between latency and reactivation/lytic replication is the regulation of the major immediate-early (MIE) locus. This enhancer/promoter region is complex, and it is regulated by chromatinization and associated factors, as well as a variety of transcription factors. Herein, we discuss these factors and how they influence the MIE locus, which ultimately impacts the phase of HCMV infection.
Collapse
|
46
|
Deng J, Huang Y, Wang Q, Li J, Ma Y, Qi Y, Liu Z, Li Y, Ruan Q. Human Cytomegalovirus Influences Host circRNA Transcriptions during Productive Infection. Virol Sin 2020; 36:241-253. [PMID: 32757146 DOI: 10.1007/s12250-020-00275-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a double-strand DNA virus widely infected in human. Circular RNAs (circRNAs) are non-coding RNAs with most functions of which keep unknown, and the effects of HCMV productive infection on host circRNA transcriptions remain unclear. In this study, we profiled 283 host circRNAs that significantly altered by HCMV productive infection in human embryonic lung fibroblasts (HELF) by RNA deep sequencing and bioinformatics analysis. Among these, circSP100, circMAP3K1, circPLEKHM1, and circTRIO were validated for their transcriptions and sequences. Furthermore, characteristics of circSP100 were investigated by RT-qPCR and northern blot. It was implied that circSP100 was produced from the sense strand of the SP100 gene containing six exons. Kinetics of circSP100 and SP100 mRNA were significantly different after infection: circSP100 levels increased gradually along with infection, whereas SP100 mRNA levels increased in the beginning and dropped at 24 h post-infection (hpi). Meanwhile, a total number of 257 proteins, including 10 HCMV encoding proteins, were identified potentially binding to cytoplasmic circSP100 by RNA antisense purification (RAP) and mass spectrometry. Enrichment analysis showed these proteins were mainly involved in the spliceosome, protein processing, ribosome, and phagosome pathways, suggesting multiple functions of circSP100 during HCMV infection.
Collapse
Affiliation(s)
- Jingui Deng
- Virology Laboratory, The Affiliated Shengjing Hospital, China Medical University, Shenyang, 110004, China.,Department of Laboratory, Central Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, China
| | - Yujing Huang
- Virology Laboratory, The Affiliated Shengjing Hospital, China Medical University, Shenyang, 110004, China.
| | - Qing Wang
- Virology Laboratory, The Affiliated Shengjing Hospital, China Medical University, Shenyang, 110004, China
| | - Jianming Li
- Virology Laboratory, The Affiliated Shengjing Hospital, China Medical University, Shenyang, 110004, China
| | - Yanping Ma
- Virology Laboratory, The Affiliated Shengjing Hospital, China Medical University, Shenyang, 110004, China
| | - Ying Qi
- Virology Laboratory, The Affiliated Shengjing Hospital, China Medical University, Shenyang, 110004, China
| | - Zhongyang Liu
- Virology Laboratory, The Affiliated Shengjing Hospital, China Medical University, Shenyang, 110004, China
| | - Yibo Li
- Department of Gynecology and Obstetrics, The Affiliated Shengjing Hospital, China Medical University, Shenyang, 110004, China.,Department of Obstetrics, Central Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, China
| | - Qiang Ruan
- Virology Laboratory, The Affiliated Shengjing Hospital, China Medical University, Shenyang, 110004, China.
| |
Collapse
|
47
|
Lau B, Kerr K, Gu Q, Nightingale K, Antrobus R, Suárez NM, Stanton RJ, Wang ECY, Weekes MP, Davison AJ. Human Cytomegalovirus Long Non-coding RNA1.2 Suppresses Extracellular Release of the Pro-inflammatory Cytokine IL-6 by Blocking NF-κB Activation. Front Cell Infect Microbiol 2020; 10:361. [PMID: 32793512 PMCID: PMC7387431 DOI: 10.3389/fcimb.2020.00361] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/11/2020] [Indexed: 12/16/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are transcripts of >200 nucleotides that are not translated into functional proteins. Cellular lncRNAs have been shown to act as regulators by interacting with target nucleic acids or proteins and modulating their activities. We investigated the role of RNA1.2, which is one of four major lncRNAs expressed by human cytomegalovirus (HCMV), by comparing the properties of parental virus in vitro with those of deletion mutants lacking either most of the RNA1.2 gene or only the TATA element of the promoter. In comparison with parental virus, these mutants exhibited no growth defects and minimal differences in viral gene expression in human fibroblasts. In contrast, 76 cellular genes were consistently up- or down-regulated by the mutants at both the RNA and protein levels at 72 h after infection. Differential expression of the gene most highly upregulated by the mutants (Tumor protein p63-regulated gene 1-like protein; TPRG1L) was confirmed at both levels by RT-PCR and immunoblotting. Consistent with the known ability of TPRG1L to upregulate IL-6 expression via NF-κB stimulation, RNA1.2 mutant-infected fibroblasts were observed to upregulate IL-6 in addition to TPRG1L. Comparable surface expression of TNF receptors and responsiveness to TNF-α in cells infected by the parental and mutant viruses indicated that activation of signaling by TNF-α is not involved in upregulation of IL-6 by the mutants. In contrast, inhibition of NF-κB activity and knockdown of TPRG1L expression reduced the extracellular release of IL-6 by RNA1.2 mutant-infected cells, thus demonstrating that upregulation of TPRG1L activates NF-κB. The levels of MCP-1 and CXCL1 transcripts were also increased in RNA1.2 mutant-infected cells, further demonstrating the presence of active NF-κB signaling. These results suggest that RNA1.2 plays a role in manipulating intrinsic NF-κB-dependent cytokine and chemokine release during HCMV infection, thereby impacting downstream immune responses.
Collapse
Affiliation(s)
- Betty Lau
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Karen Kerr
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Quan Gu
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Katie Nightingale
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Nicolás M. Suárez
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Richard J. Stanton
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Eddie C. Y. Wang
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael P. Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Andrew J. Davison
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| |
Collapse
|
48
|
Chaturvedi S, Klein J, Vardi N, Bolovan-Fritts C, Wolf M, Du K, Mlera L, Calvert M, Moorman NJ, Goodrum F, Huang B, Weinberger LS. A molecular mechanism for probabilistic bet hedging and its role in viral latency. Proc Natl Acad Sci U S A 2020; 117:17240-17248. [PMID: 32632017 PMCID: PMC7382263 DOI: 10.1073/pnas.1914430117] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Probabilistic bet hedging, a strategy to maximize fitness in unpredictable environments by matching phenotypic variability to environmental variability, is theorized to account for the evolution of various fate-specification decisions, including viral latency. However, the molecular mechanisms underlying bet hedging remain unclear. Here, we report that large variability in protein abundance within individual herpesvirus virion particles enables probabilistic bet hedging between viral replication and latency. Superresolution imaging of individual virions of the human herpesvirus cytomegalovirus (CMV) showed that virion-to-virion levels of pp71 tegument protein-the major viral transactivator protein-exhibit extreme variability. This super-Poissonian tegument variability promoted alternate replicative strategies: high virion pp71 levels enhance viral replicative fitness but, strikingly, impede silencing, whereas low virion pp71 levels reduce fitness but promote silencing. Overall, the results indicate that stochastic tegument packaging provides a mechanism enabling probabilistic bet hedging between viral replication and latency.
Collapse
Affiliation(s)
- Sonali Chaturvedi
- Gladstone Institute for Virology and Immunology, Gladstone|University of California, San Francisco Center for Cell Circuitry, San Francisco, CA 94158
| | - Jonathan Klein
- Gladstone Institute for Virology and Immunology, Gladstone|University of California, San Francisco Center for Cell Circuitry, San Francisco, CA 94158
| | - Noam Vardi
- Gladstone Institute for Virology and Immunology, Gladstone|University of California, San Francisco Center for Cell Circuitry, San Francisco, CA 94158
| | - Cynthia Bolovan-Fritts
- Gladstone Institute for Virology and Immunology, Gladstone|University of California, San Francisco Center for Cell Circuitry, San Francisco, CA 94158
| | - Marie Wolf
- Gladstone Institute for Virology and Immunology, Gladstone|University of California, San Francisco Center for Cell Circuitry, San Francisco, CA 94158
| | - Kelvin Du
- Gladstone Institute for Virology and Immunology, Gladstone|University of California, San Francisco Center for Cell Circuitry, San Francisco, CA 94158
| | - Luwanika Mlera
- Department of Cell & Molecular Medicine, University of Arizona, Tucson, AZ 85721
| | - Meredith Calvert
- Gladstone Institute for Virology and Immunology, Gladstone|University of California, San Francisco Center for Cell Circuitry, San Francisco, CA 94158
| | - Nathaniel J Moorman
- Department of Microbiology & Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Felicia Goodrum
- Department of Cell & Molecular Medicine, University of Arizona, Tucson, AZ 85721
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158
| | - Leor S Weinberger
- Gladstone Institute for Virology and Immunology, Gladstone|University of California, San Francisco Center for Cell Circuitry, San Francisco, CA 94158;
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158
| |
Collapse
|
49
|
Poole E, Sinclair J. Understanding HCMV Latency Using Unbiased Proteomic Analyses. Pathogens 2020; 9:E590. [PMID: 32698381 PMCID: PMC7399836 DOI: 10.3390/pathogens9070590] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
Human cytomegalovirus (HCMV) establishes either a latent (non-productive) or lytic (productive) infection depending upon cell type, cytokine milieu and the differentiation status of the infected cell. Undifferentiated cells, such as precursor cells of the myeloid lineage, support a latent infection whereas terminally differentiated cells, such as monocytes or dendritic cells are an environment conducive to reactivation and support a lytic infection. The mechanisms which regulate HCMV in either a latent or lytic infection have been the focus of intense investigation with a view to developing novel treatments for HCMV-associated disease which can have a heavy clinical burden after reactivation or primary infection in, especially, the immune compromised. To this end, a number of studies have been carried out in an unbiased manner to address global changes occurring within the latently infected cell to address the molecular changes associated with HCMV latency. In this review, we will concentrate on the proteomic analyses which have been carried out in undifferentiated myeloid cells which either stably express specific viral latency associated genes in isolation or on cells which have been latently infected with virus.
Collapse
Affiliation(s)
- Emma Poole
- Department of Medicine, University of Cambridge, box 157, Level 5 Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, UK;
| | | |
Collapse
|
50
|
Wang YQ, Zhao XY. Human Cytomegalovirus Primary Infection and Reactivation: Insights From Virion-Carried Molecules. Front Microbiol 2020; 11:1511. [PMID: 32765441 PMCID: PMC7378892 DOI: 10.3389/fmicb.2020.01511] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV), a ubiquitous beta-herpesvirus, is able to establish lifelong latency after initial infection. Periodical reactivation occurs after immunosuppression, remaining a major cause of death in immunocompromised patients. HCMV has to reach a structural and functional balance with the host at its earliest entry. Virion-carried mediators are considered to play pivotal roles in viral adaptation into a new cellular environment upon entry. Additionally, one clear difference between primary infection and reactivation is the idea that virion-packaged factors are already formed such that those molecules can be used swiftly by the virus. In contrast, virion-carried mediators have to be transcribed and translated; thus, they are not readily available during reactivation. Hence, understanding virion-carried molecules helps to elucidate HCMV reactivation. In this article, the impact of virion-packaged molecules on viral structure, biological behavior, and viral life cycle will be reviewed.
Collapse
Affiliation(s)
- Yu-Qing Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,PKU-THU Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| |
Collapse
|