1
|
Herb S, Zeleznjak J, Hennig T, L'Hernault A, Lodha M, Jürges C, Trsan T, Juranic Lisnic V, Jonjic S, Erhard F, Krmpotic A, Dölken L. Two murine cytomegalovirus microRNAs target the major viral immediate early 3 gene. J Gen Virol 2022; 103. [DOI: 10.1099/jgv.0.001804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Human cytomegalovirus is responsible for morbidity and mortality in immune compromised patients and is the leading viral cause of congenital infection. Virus-encoded microRNAs (miRNAs) represent interesting targets for novel antiviral agents. While many cellular targets that augment productive infection have been identified in recent years, regulation of viral genes such as the major viral immediate early protein 72 (IE72) by hcmv-miR-UL112-1 may contribute to both the establishment and the maintenance of latent infection. We employed photoactivated ribonucleotide-enhanced individual nucleotide resolution crosslinking (PAR-iCLIP) to identify murine cytomegalovirus (MCMV) miRNA targets during lytic infection. While the PAR-iCLIP data were of insufficient quality to obtain a comprehensive list of cellular and viral miRNA targets, the most prominent PAR-iCLIP peak in the MCMV genome mapped to the 3′ untranslated region of the major viral immediate early 3 (ie3) transcript. We show that this results from two closely positioned binding sites for the abundant MCMV miRNAs miR-M23-2-3p and miR-m01-2-3p. Their pre-expression significantly impaired viral plaque formation. However, mutation of the respective binding sites did not alter viral fitness during acute or subacute infection in vivo. Furthermore, no differences in the induction of virus-specific CD8+ T cells were observed. Future studies will probably need to go beyond studying immunocompetent laboratory mice housed in pathogen-free conditions to reveal the functional relevance of viral miRNA-mediated regulation of key viral immediate early genes.
Collapse
Affiliation(s)
- Stefanie Herb
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
| | - Jelena Zeleznjak
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Thomas Hennig
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
| | - Anne L'Hernault
- Department of Medicine, University of Cambridge, Box 157, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Manivel Lodha
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
| | - Christopher Jürges
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
| | - Tihana Trsan
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Vanda Juranic Lisnic
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Stipan Jonjic
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Florian Erhard
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
| | - Astrid Krmpotic
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Lars Dölken
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), 97080 Würzburg, Germany
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
- Department of Medicine, University of Cambridge, Box 157, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
2
|
Afshari A, Yaghobi R, Rezaei G. Inter-regulatory role of microRNAs in interaction between viruses and stem cells. World J Stem Cells 2021; 13:985-1004. [PMID: 34567421 PMCID: PMC8422934 DOI: 10.4252/wjsc.v13.i8.985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/11/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are well known for post-transcriptional regulatory ability over specific mRNA targets. miRNAs exhibit temporal or tissue-specific expression patterns and regulate the cell and tissue developmental pathways. They also have determinative roles in production and differentiation of multiple lineages of stem cells and might have therapeutic advantages. miRNAs are a part of some viruses’ regulatory machinery, not a byproduct. The trace of miRNAs was detected in the genomes of viruses and regulation of cell reprograming and viral pathogenesis. Combination of inter-regulatory systems has been detected for miRNAs during viral infections in stem cells. Contraction between viruses and stem cells may be helpful in therapeutic tactics, pathogenesis, controlling viral infections and defining stem cell developmental strategies that is programmed by miRNAs as a tool. Therefore, in this review we intended to study the inter-regulatory role of miRNAs in the interaction between viruses and stem cells and tried to explain the advantages of miRNA regulatory potentials, which make a new landscape for future studies.
Collapse
Affiliation(s)
- Afsoon Afshari
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz 7193711351, Iran
| | - Ramin Yaghobi
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz 7193711351, Iran
| | - Ghazal Rezaei
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz 7193711351, Iran
| |
Collapse
|
3
|
Chan B, Arapović M, Masters LL, Rwandamuiye F, Jonjić S, Smith LM, Redwood AJ. The m15 Locus of Murine Cytomegalovirus Modulates Natural Killer Cell Responses to Promote Dissemination to the Salivary Glands and Viral Shedding. Pathogens 2021; 10:pathogens10070866. [PMID: 34358016 PMCID: PMC8308470 DOI: 10.3390/pathogens10070866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 11/16/2022] Open
Abstract
As the largest herpesviruses, the 230 kb genomes of cytomegaloviruses (CMVs) have increased our understanding of host immunity and viral escape mechanisms, although many of the annotated genes remain as yet uncharacterised. Here we identify the m15 locus of murine CMV (MCMV) as a viral modulator of natural killer (NK) cell immunity. We show that, rather than discrete transcripts from the m14, m15 and m16 genes as annotated, there are five 3′-coterminal transcripts expressed over this region, all utilising a consensus polyA tail at the end of the m16 gene. Functional inactivation of any one of these genes had no measurable impact on viral replication. However, disruption of all five transcripts led to significantly attenuated dissemination to, and replication in, the salivary glands of multiple strains of mice, but normal growth during acute infection. Disruption of the m15 locus was associated with heightened NK cell responses, including enhanced proliferation and IFNγ production. Depletion of NK cells, but not T cells, rescued salivary gland replication and viral shedding. These data demonstrate the identification of multiple transcripts expressed by a single locus which modulate, perhaps in a concerted fashion, the function of anti-viral NK cells.
Collapse
Affiliation(s)
- Baca Chan
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (B.C.); (L.L.M.); (F.R.); (L.M.S.)
- Institute of Respiratory Health, University of Western Australia, Nedlands, WA 6009, Australia
| | - Maja Arapović
- Department for Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (M.A.); (S.J.)
| | - Laura L. Masters
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (B.C.); (L.L.M.); (F.R.); (L.M.S.)
| | - Francois Rwandamuiye
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (B.C.); (L.L.M.); (F.R.); (L.M.S.)
| | - Stipan Jonjić
- Department for Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (M.A.); (S.J.)
| | - Lee M. Smith
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (B.C.); (L.L.M.); (F.R.); (L.M.S.)
| | - Alec J. Redwood
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (B.C.); (L.L.M.); (F.R.); (L.M.S.)
- Institute of Respiratory Health, University of Western Australia, Nedlands, WA 6009, Australia
- Correspondence: ; Tel.: +61-8-6151-0895
| |
Collapse
|
4
|
Lee BJ, Min CK, Hancock M, Streblow DN, Caposio P, Goodrum FD, Yurochko AD. Human Cytomegalovirus Host Interactions: EGFR and Host Cell Signaling Is a Point of Convergence Between Viral Infection and Functional Changes in Infected Cells. Front Microbiol 2021; 12:660901. [PMID: 34025614 PMCID: PMC8138183 DOI: 10.3389/fmicb.2021.660901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/07/2021] [Indexed: 12/22/2022] Open
Abstract
Viruses have evolved diverse strategies to manipulate cellular signaling pathways in order to promote infection and/or persistence. Human cytomegalovirus (HCMV) possesses a number of unique properties that allow the virus to alter cellular events required for infection of a diverse array of host cell types and long-term persistence. Of specific importance is infection of bone marrow derived and myeloid lineage cells, such as peripheral blood monocytes and CD34+ hematopoietic progenitor cells (HPCs) because of their essential role in dissemination of the virus and for the establishment of latency. Viral induced signaling through the Epidermal Growth Factor Receptor (EGFR) and other receptors such as integrins are key control points for viral-induced cellular changes and productive and latent infection in host organ systems. This review will explore the current understanding of HCMV strategies utilized to hijack cellular signaling pathways, such as EGFR, to promote the wide-spread dissemination and the classic life-long herpesvirus persistence.
Collapse
Affiliation(s)
- Byeong-Jae Lee
- Department of Microbiology & Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center of Excellence for Emerging Viral Threats, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
| | - Chan-Ki Min
- Department of Microbiology & Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center of Excellence for Emerging Viral Threats, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
| | - Meaghan Hancock
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| | | | - Andrew D Yurochko
- Department of Microbiology & Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center of Excellence in Arthritis and Rheumatology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
| |
Collapse
|
5
|
Abstract
The "omics" revolution of recent years has simplified the study of RNA transcripts produced during viral infection and under specific defined conditions. In the quest to find new and differentially expressed transcripts during the course of human Herpesvirus 6B (HHV-6B) infection, we made use of large-scale RNA sequencing to analyze the HHV-6B transcriptome during productive infection of human Molt-3 T-cells. Analyses were performed at different time points following infection and specific inhibitors were used to classify the kinetic class of each open reading frame (ORF) reported in the annotated genome of HHV-6B Z29 strain. The initial search focussed on HHV-6B-specific reads matching new HHV-6B transcripts. Differential expression of new HHV-6B transcripts were observed in all samples analyzed. The presence of many of these new HHV-6B transcripts were confirmed by RT-PCR and Sanger sequencing. Many of these transcripts represented new splice variants of previously reported ORFs, including some transcripts that have yet to be defined. Overall, our work demonstrates the diversity and the complexity of the HHV-6B transcriptome.IMPORTANCERNA sequencing (RNA-seq) is an important tool for studying RNA transcripts, particularly during active viral infection. We made use of RNA-seq to study human Herpesvirus 6B (HHV-6B) infection. Using six different time points, we were able to identify the presence of differentially spliced genes at 6, 9, 12, 24, 48 and 72 hours post-infection. Determination of the RNA profiles in the presence of cycloheximide (CHX) or phosphonoacetic acid (PAA) also permitted identification of the kinetic class of each ORF described in the annotated GenBank file. We also identified new spliced transcripts for certain genes and evaluated their relative expression over time. These data and next-generation sequencing (NGS) of the viral DNA have led us to propose a new version of the HHV-6B Z29 GenBank annotated file, without changing ORF names in order to facilitate trace back and correlate our work with previous studies on HHV-6B.
Collapse
|
6
|
Luo Y, Yu L, Feng Z, Chen Q, Lu L, Zhang Q, Xu D. Integrated analysis of viral miRNAs, mRNA and protein in the caudal fin cells of C. auratus gibelio with cyprinid herpesvirus 2 infection. JOURNAL OF FISH DISEASES 2021; 44:441-460. [PMID: 33577719 DOI: 10.1111/jfd.13289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/26/2020] [Accepted: 09/29/2020] [Indexed: 06/12/2023]
Abstract
Cyprinid herpesvirus 2 (CyHV-2), a member of the genus Cyprinivirus in the family Alloherpesviridae, has attracted worldwide attention because it causes severe disease and high mortality in crucian carp and goldfish. In this study, we focus on mRNA, protein and viral miRNA expression profiles in C. auratus gibelio caudal fin (GiCF) cells infected with CyHV-2, using high-throughput sequence techniques and TMT-labelled analyses. The results revealed that 156 virus genes were differentially expressed during the infection. Among these differentially expressed genes, 7 viral genes were significantly up-regulated and 28 were significantly down-regulated at 96 hpi (hours post-infection) vs 48 hpi. Besides, a total of 78 viral proteins, including a large number of membrane proteins and capsid proteins associated with the viral assembly, were successfully detected by using proteome analysis. Furthermore, a total of 225,143,474 raw reads were generated from cDNA library of CyHV-2-infected GiCF cells using high-throughput sequencing technology. Following annotation and secondary structure prediction, 10 viral miRNAs were found as significantly modulated in CyHV-2-infected GiCF cells (2 down-regulated and 8 up-regulated). Finally, the CyHV-2 genes (orf19, orf23, orf118, orf121, orf127) targeted by the viral miRNA CyHV-2-KT-635 identified in this study, were predicted and validated by quantitative reverse transcription polymerase chain reaction (qRT-PCR), and the regulation of CyHV-2-KT-635 on orf121 protein expression was verified by western blotting assay. Taken together, this study provides a valuable basis for further research on the expression of virus genes during CyHV-2 replication and the molecular mechanisms by which miRNA may regulate CyHV-2 virus.
Collapse
Affiliation(s)
- Yang Luo
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China
| | - Lu Yu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China
| | - Zizhao Feng
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China
| | - Qikang Chen
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China
| | - Liqun Lu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China
- National Experimental Teaching Demonstration Center for Fishery Sciences, Shanghai Ocean University, Shanghai, China
| | - Qiya Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Wuhan, China
| | - Dan Xu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, China
- National Experimental Teaching Demonstration Center for Fishery Sciences, Shanghai Ocean University, Shanghai, China
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Sciences, Nanning, China
| |
Collapse
|
7
|
Diggins NL, Crawford LB, Struthers HM, Hook LM, Landais I, Skalsky RL, Hancock MH. Techniques for Characterizing Cytomegalovirus-Encoded miRNAs. Methods Mol Biol 2021; 2244:301-342. [PMID: 33555594 DOI: 10.1007/978-1-0716-1111-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
microRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at the posttranscriptional level by binding to sites within the 3' untranslated regions of messenger RNA (mRNA) transcripts. The discovery of this completely new mechanism of gene regulation necessitated the development of a variety of techniques to further characterize miRNAs, their expression, and function. In this chapter, we will discuss techniques currently used in the miRNA field to detect, express and inhibit miRNAs, as well as methods used to identify and validate their targets, specifically with respect to the miRNAs encoded by human cytomegalovirus.
Collapse
Affiliation(s)
- Nicole L Diggins
- Vaccine and Gene Therapy Institute, Oregon Health and Sciences University, Beaverton, OR, USA
| | - Lindsey B Crawford
- Vaccine and Gene Therapy Institute, Oregon Health and Sciences University, Beaverton, OR, USA
| | - Hillary M Struthers
- Vaccine and Gene Therapy Institute, Oregon Health and Sciences University, Beaverton, OR, USA
| | - Lauren M Hook
- Vaccine and Gene Therapy Institute, Oregon Health and Sciences University, Beaverton, OR, USA
| | - Igor Landais
- Vaccine and Gene Therapy Institute, Oregon Health and Sciences University, Beaverton, OR, USA
| | - Rebecca L Skalsky
- Vaccine and Gene Therapy Institute, Oregon Health and Sciences University, Beaverton, OR, USA
| | - Meaghan H Hancock
- Vaccine and Gene Therapy Institute, Oregon Health and Sciences University, Beaverton, OR, USA.
| |
Collapse
|
8
|
Affiliation(s)
- Lucy Ginn
- Transcriptional Networks in Lung Cancer Group Cancer Research UK Manchester Institute University of Manchester Manchester UK
- Cancer Research UK Lung Cancer Centre of Excellence At Manchester and University College London England UK
| | - Manuela La Montagna
- Transcriptional Networks in Lung Cancer Group Cancer Research UK Manchester Institute University of Manchester Manchester UK
- Cancer Research UK Lung Cancer Centre of Excellence At Manchester and University College London England UK
| | - Qinghua Wu
- College of Life Science Yangtze University Jingzhou Hubei China
- Department of Chemistry Faculty of Science University of Hradec Kralove Hradec Kralove East Bohemia Czech Republic
| | - Lei Shi
- Transcriptional Networks in Lung Cancer Group Cancer Research UK Manchester Institute University of Manchester Manchester UK
- Cancer Research UK Lung Cancer Centre of Excellence At Manchester and University College London England UK
| |
Collapse
|
9
|
Mendes AF, Goncalves P, Serrano-Solis V, Silva PMD. Identification of candidate microRNAs from Ostreid herpesvirus-1 (OsHV-1) and their potential role in the infection of Pacific oysters (Crassostrea gigas). Mol Immunol 2020; 126:153-164. [PMID: 32853878 DOI: 10.1016/j.molimm.2020.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 12/21/2022]
Abstract
Oyster production is an economic activity of great interest worldwide. Recently, oysters have been suffering significant mortalities from OsHV-1infection, which has resulted in substantial economic loses in several countries around the world. Understanding viral pathogenicity mechanisms is of central importance for the establishment of disease control measures. Thus, the present work aimed to identify and characterize miRNAs from OsHV-1 as well as to predict their target transcripts in the virus and the host. OsHV-1 genome was used for the in silico discovery of pre-miRNAs. Subsequently, viral and host target transcripts of the OsHV-1 miRNAs were predicted according to the base pairing interaction between mature miRNAs and mRNA 3' untranslated regions (UTRs). Six unique pre-miRNAs were found in different regions of the viral genome, ranging in length from 85 to 172 nucleotides. A complex network of self-regulation of viral gene expression mediated by the miRNAs was identified. These sequences also seem to have a broad ability to regulate the expression of host immune-related genes, especially those associated with pathogen recognition. Our results suggest that OsHV-1 encodes miRNAs with important functions in the infection process, inducing self-regulation of viral transcripts, as well as affecting the regulation of Pacific oyster transcripts related to immunity. Understanding the molecular basis of host-pathogen interactions can help mitigate the recurrent events of oyster mass mortalities by OsHV-1 observed worldwide.
Collapse
Affiliation(s)
- Andrei Félix Mendes
- Laboratório de Imunologia e Patologia de Invertebrados (LABIPI), Departamento de Biologia Molecular, Universidade Federal da Paraíba (UFPB), 58051-900, João Pessoa, Paraíba, Brazil
| | - Priscila Goncalves
- Environment and Sustainability Institute, University of Exeter, Penryn, Cornwall, TR10 9FE, UK
| | - Victor Serrano-Solis
- Laboratório de Imunologia e Patologia de Invertebrados (LABIPI), Departamento de Biologia Molecular, Universidade Federal da Paraíba (UFPB), 58051-900, João Pessoa, Paraíba, Brazil
| | - Patricia Mirella da Silva
- Laboratório de Imunologia e Patologia de Invertebrados (LABIPI), Departamento de Biologia Molecular, Universidade Federal da Paraíba (UFPB), 58051-900, João Pessoa, Paraíba, Brazil.
| |
Collapse
|
10
|
Zhang L, Yu J, Liu Z. MicroRNAs expressed by human cytomegalovirus. Virol J 2020; 17:34. [PMID: 32164742 PMCID: PMC7069213 DOI: 10.1186/s12985-020-1296-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
Background MicroRNAs (miRNAs) are small non-coding RNAs about 22 nucleotides in length, which play an important role in gene regulation of both eukaryotes and viruses. They can promote RNA cleavage and repress translation via base-pairing with complementary sequences within mRNA molecules. Main body Human cytomegalovirus (HCMV) encodes a large number of miRNAs that regulate transcriptions of both host cells and themselves to favor viral infection and inhibit the host’s immune response. To date, ~ 26 mature HCMV miRNAs have been identified. Nevertheless, their roles in viral infection are ambiguous, and the mechanisms have not been fully revealed. Therefore, we discuss the methods used in HCMV miRNA research and summarize the important roles of HCMV miRNAs and their potential mechanisms in infection. Conclusions To study the miRNAs encoded by viruses and their roles in viral replication, expression, and infection will not only contribute to the planning of effective antiviral therapies, but also provide new molecular targets for the development of antiviral drugs.
Collapse
Affiliation(s)
- Lichen Zhang
- Clinical School, Weifang Medical University, Weifang, 261053, China
| | - Jiaqi Yu
- Clinical School, Weifang Medical University, Weifang, 261053, China
| | - Zhijun Liu
- Department of Medical Microbiology, Weifang Medical University, Weifang, 261053, China.
| |
Collapse
|
11
|
Hancock MH, Skalsky RL. Roles of Non-coding RNAs During Herpesvirus Infection. Curr Top Microbiol Immunol 2019; 419:243-280. [PMID: 28674945 DOI: 10.1007/82_2017_31] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Non-coding RNAs (ncRNAs) play essential roles in multiple aspects of the life cycles of herpesviruses and contribute to lifelong persistence of herpesviruses within their respective hosts. In this chapter, we discuss the types of ncRNAs produced by the different herpesvirus families during infection, some of the cellular ncRNAs manipulated by these viruses, and the overall contributions of ncRNAs to the viral life cycle, influence on the host environment, and pathogenesis.
Collapse
Affiliation(s)
- Meaghan H Hancock
- Vaccine and Gene Therapy Institute at Oregon Health and Science University, Beaverton, OR, USA
| | - Rebecca L Skalsky
- Vaccine and Gene Therapy Institute at Oregon Health and Science University, Beaverton, OR, USA.
| |
Collapse
|
12
|
Herpes Simplex Virus 1 Lytic Infection Blocks MicroRNA (miRNA) Biogenesis at the Stage of Nuclear Export of Pre-miRNAs. mBio 2019; 10:mBio.02856-18. [PMID: 30755517 PMCID: PMC6372804 DOI: 10.1128/mbio.02856-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Various mechanisms have been identified by which viruses target host small RNA biogenesis pathways to achieve optimal infection outcomes. Herpes simplex virus 1 (HSV-1) is a ubiquitous human pathogen whose successful persistence in the host entails both productive (“lytic”) and latent infection. Although many HSV-1 miRNAs have been discovered and some are thought to help control the lytic/latent switch, little is known about regulation of their biogenesis. By characterizing expression of both pre-miRNAs and mature miRNAs under various conditions, this study revealed striking differences in miRNA biogenesis between lytic and latent infection and uncovered a regulatory mechanism that blocks pre-miRNA nuclear export and is dependent on viral protein ICP27 and viral DNA synthesis. This mechanism represents a new virus-host interaction that could limit the repressive effects of HSV-1 miRNAs hypothesized to promote latency and may shed light on the regulation of miRNA nuclear export, which has been relatively unexplored. Herpes simplex virus 1 (HSV-1) switches between two infection programs, productive (“lytic”) and latent infection. Some HSV-1 microRNAs (miRNAs) have been hypothesized to help control this switch, and yet little is known about regulation of their expression. Using Northern blot analyses, we found that, despite inherent differences in biogenesis efficiency among six HSV-1 miRNAs, all six exhibited high pre-miRNA/miRNA ratios during lytic infection of different cell lines and, when detectable, in acutely infected mouse trigeminal ganglia. In contrast, considerably lower ratios were observed in latently infected ganglia and in cells transduced with lentiviral vectors expressing the miRNAs, suggesting that HSV-1 lytic infection blocks miRNA biogenesis. This phenomenon is not specific to viral miRNAs, as a host miRNA expressed from recombinant HSV-1 also exhibited high pre-miRNA/miRNA ratios late during lytic infection. The levels of most of the mature miRNAs remained stable during infection in the presence of actinomycin D, indicating that the high ratios are due to inefficient pre-miRNA conversion to miRNA. Cellular fractionation experiments showed that late (but not early) during infection, pre-miRNAs were enriched in the nucleus and depleted in the cytoplasm, indicating that nuclear export was blocked. A mutation eliminating ICP27 expression or addition of acyclovir reduced pre-miRNA/miRNA ratios, but mutations drastically reducing Us11 expression did not. Thus, HSV-1 lytic infection inhibits miRNA biogenesis at the step of nuclear export and does so in an ICP27- and viral DNA synthesis-dependent manner. This mechanism may benefit the virus by reducing expression of repressive miRNAs during lytic infection while permitting elevated expression during latency.
Collapse
|
13
|
HCMV miRNA Targets Reveal Important Cellular Pathways for Viral Replication, Latency, and Reactivation. Noncoding RNA 2018; 4:ncrna4040029. [PMID: 30360396 PMCID: PMC6315856 DOI: 10.3390/ncrna4040029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/12/2018] [Accepted: 10/17/2018] [Indexed: 02/06/2023] Open
Abstract
It is now well appreciated that microRNAs (miRNAs) play a critical role in the lifecycles of many herpes viruses. The human cytomegalovirus (HCMV) replication cycle varies significantly depending on the cell type infected, with lytic replication occurring in fully-differentiated cells such as fibroblasts, endothelial cells, or macrophages, and latent infection occurring in less-differentiated CD14+ monocytes and CD34+ hematopoietic progenitor cells where viral gene expression is severely diminished and progeny virus is not produced. Given their non-immunogenic nature and their capacity to target numerous cellular and viral transcripts, miRNAs represent a particularly advantageous means for HCMV to manipulate viral gene expression and cellular signaling pathways during lytic and latent infection. This review will focus on our current knowledge of HCMV miRNA viral and cellular targets, and discuss their importance in lytic and latent infection, highlight the challenges of studying HCMV miRNAs, and describe how viral miRNAs can help us to better understand the cellular processes involved in HCMV latency.
Collapse
|
14
|
Sprague L, Braidwood L, Conner J, Cassady KA, Benencia F, Cripe TP. Please stand by: how oncolytic viruses impact bystander cells. Future Virol 2018; 13:671-680. [PMID: 30416535 DOI: 10.2217/fvl-2018-0068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 05/23/2018] [Indexed: 12/19/2022]
Abstract
Oncolytic viruses (OVs) do more than simply infect and kill host cells. The accepted mechanism of action for OVs consists of a primary lytic phase and a subsequent antitumor and antiviral immune response. However, not all cells are subject to the direct effects of OV therapy, and it is becoming clear that OVs can also impact uninfected cells in the periphery. This review discusses the effects of OVs on uninfected neighboring cells, so-called bystander effects, and implications for OV therapies alone or in combination with other standard of care chemotherapy.
Collapse
Affiliation(s)
- Leslee Sprague
- The Ohio State University College of Medicine, Biomedical Sciences Graduate Program, Columbus, 43201 OH, USA.,The Ohio State University College of Medicine, Biomedical Sciences Graduate Program, Columbus, 43201 OH, USA
| | - Lynne Braidwood
- Virttu Biologics, BioCity, Scotland, UK.,Virttu Biologics, BioCity, Scotland, UK
| | - Joe Conner
- Virttu Biologics, BioCity, Scotland, UK.,Virttu Biologics, BioCity, Scotland, UK
| | - Kevin A Cassady
- Nationwide Children's Hospital, Division of Infectious Diseases, Columbus, 43205 OH, USA.,Nationwide Children's Hospital, Division of Hematology/Oncology/BMT & Center for Childhood Cancer & Blood Diseases, Columbus, 43205 OH, USA.,Nationwide Children's Hospital, Division of Infectious Diseases, Columbus, 43205 OH, USA.,Nationwide Children's Hospital, Division of Hematology/Oncology/BMT & Center for Childhood Cancer & Blood Diseases, Columbus, 43205 OH, USA
| | - Fabian Benencia
- Ohio University Russ College of Engineering & Technology, Biomedical Engineering, Athens, 45701 OH, USA.,Ohio University Russ College of Engineering & Technology, Biomedical Engineering, Athens, 45701 OH, USA
| | - Timothy P Cripe
- The Ohio State University College of Medicine, Biomedical Sciences Graduate Program, Columbus, 43201 OH, USA.,Nationwide Children's Hospital, Division of Hematology/Oncology/BMT & Center for Childhood Cancer & Blood Diseases, Columbus, 43205 OH, USA.,The Ohio State University College of Medicine, Biomedical Sciences Graduate Program, Columbus, 43201 OH, USA.,Nationwide Children's Hospital, Division of Hematology/Oncology/BMT & Center for Childhood Cancer & Blood Diseases, Columbus, 43205 OH, USA
| |
Collapse
|
15
|
Lu J, Xu D, Jiang Y, Kong S, Shen Z, Xia S, Lu L. Integrated analysis of mRNA and viral miRNAs in the kidney of Carassius auratus gibelio response to cyprinid herpesvirus 2. Sci Rep 2017; 7:13787. [PMID: 29062054 PMCID: PMC5653811 DOI: 10.1038/s41598-017-14217-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 10/06/2017] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding single stranded RNAs that play crucial roles in numerous biological processes. Vertebrate herpesviruses encode multiple viral miRNAs that modulate host and viral genes. However, the roles of viral miRNAs in lower vertebrates have not been fully determined. Here, we used high-throughput sequencing to analyse the miRNA and mRNA expression profiles of Carassius auratus gibelio in response to infection by cyprinid herpesvirus 2 (CyHV-2). RNA sequencing obtained 26,664 assembled transcripts, including 2,912 differentially expressed genes. Based on small RNA sequencing and secondary structure predictions, we identified 17 CyHV-2 encoded miRNAs, among which 14 were validated by stem-loop quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) and eight were validated by northern blotting. Furthermore, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of miRNAs-mRNA pairs revealed diverse affected immune signalling pathways, including the RIG-I-like receptor and JAK-STAT pathways. Finally, we presented four genes involved in RIG-I-like pathways, including host gene IRF3, RBMX, PIN1, viral gene ORF4, which are negatively regulated by CyHV-2 encoded miRNA miR-C4. The present study is the first to provide a comprehensive overview of viral miRNA-mRNA co-regulation, which might have a key role in controlling post-transcriptomic regulation during CyHV-2 infection.
Collapse
Affiliation(s)
- Jianfei Lu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, P. R. China
| | - Dan Xu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, P. R. China
- Key Laboratory of Agriculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, P. R. China
- National Experimental Teaching Demonstration Center for Fishery Sciences, Shanghai Ocean University, Shanghai, P. R. China
| | - Yousheng Jiang
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, P. R. China
- Key Laboratory of Agriculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, P. R. China
- National Experimental Teaching Demonstration Center for Fishery Sciences, Shanghai Ocean University, Shanghai, P. R. China
| | - Shanyun Kong
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, P. R. China
| | - Zhaoyuan Shen
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, P. R. China
| | - Siyao Xia
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, P. R. China
| | - Liqun Lu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, P. R. China.
- Key Laboratory of Agriculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, P. R. China.
- National Experimental Teaching Demonstration Center for Fishery Sciences, Shanghai Ocean University, Shanghai, P. R. China.
| |
Collapse
|
16
|
Kim H, Iizasa H, Kanehiro Y, Fekadu S, Yoshiyama H. Herpesviral microRNAs in Cellular Metabolism and Immune Responses. Front Microbiol 2017; 8:1318. [PMID: 28769892 PMCID: PMC5513955 DOI: 10.3389/fmicb.2017.01318] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/29/2017] [Indexed: 12/15/2022] Open
Abstract
The microRNAs (miRNAs) function as a key regulator in many biological processes through post-transcriptional suppression of messenger RNAs. Recent advancements have revealed that miRNAs are involved in many biological functions of cells. Not only host cells, but also some viruses encode miRNAs in their genomes. Viral miRNAs regulate cell proliferation, differentiation, apoptosis, and the cell cycle to establish infection and produce viral progeny. Particularly, miRNAs encoded by herpes virus families play integral roles in persistent viral infection either by regulation of metabolic processes or the immune response of host cells. The life-long persistent infection of gamma herpes virus subfamilies, such as Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus, induces host cells to malignant transformation. The unbalanced metabolic processes and evasion from host immune surveillance by viral miRNAs are induced either by direct targeting of key proteins or indirect regulation of multiple signaling pathways. We provide an overview of the pathogenic roles of viral miRNAs in cellular metabolism and immune responses during herpesvirus infection.
Collapse
Affiliation(s)
- Hyoji Kim
- Department of Microbiology, Faculty of Medicine, Shimane UniversityShimane, Japan
| | - Hisashi Iizasa
- Department of Microbiology, Faculty of Medicine, Shimane UniversityShimane, Japan
| | - Yuichi Kanehiro
- Department of Microbiology, Faculty of Medicine, Shimane UniversityShimane, Japan
| | - Sintayehu Fekadu
- Department of Microbiology, Faculty of Medicine, Shimane UniversityShimane, Japan
| | - Hironori Yoshiyama
- Department of Microbiology, Faculty of Medicine, Shimane UniversityShimane, Japan
| |
Collapse
|
17
|
Jiang S, Qi Y, He R, Huang Y, Liu Z, Ma Y, Guo X, Shao Y, Sun Z, Ruan Q. Human cytomegalovirus microRNA miR-US25-1-5p inhibits viral replication by targeting multiple cellular genes during infection. Gene 2015; 570:108-14. [PMID: 26055091 DOI: 10.1016/j.gene.2015.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/29/2015] [Accepted: 06/03/2015] [Indexed: 11/28/2022]
Abstract
MicroRNAs (miRNAs) play important roles in regulating various cellular processes in plants, animals, and viruses. This mechanism is also utilized by human cytomegalovirus (HCMV) in the process of infection and pathogenesis. The HCMV-encoded miRNA, hcmv-miR-US25-1-5p, was highly expressed during lytic and latent infections, and was found to inhibit viral replication. Identification of functional target genes of this microRNA is important in that it will enable a better understanding of the function of hcmv-miR-US25-1-5p during HCMV infection. In the present study, 35 putative cellular transcript targets of hcmv-miR-US25-1-5p were identified. Down-regulation of the targets YWHAE, UBB, NPM1, and HSP90AA1 by hcmv-miR-US25-1-5p was validated by luciferase reporter assay and Western blot analysis. In addition, we showed that hcmv-miR-US25-1-5p could inhibit viral replication by interacting with these targets, the existence of which may impact virus replication directly or indirectly.
Collapse
Affiliation(s)
- Shujuan Jiang
- Virus Laboratory, The Affiliated Shengjing Hospital, China Medical University, China; Clinical Genetics, The Affiliated Shengjing Hospital, China Medical University, China
| | - Ying Qi
- Virus Laboratory, The Affiliated Shengjing Hospital, China Medical University, China
| | - Rong He
- Clinical Genetics, The Affiliated Shengjing Hospital, China Medical University, China.
| | - Yujing Huang
- Virus Laboratory, The Affiliated Shengjing Hospital, China Medical University, China
| | - Zhongyang Liu
- Virus Laboratory, The Affiliated Shengjing Hospital, China Medical University, China
| | - Yanping Ma
- Virus Laboratory, The Affiliated Shengjing Hospital, China Medical University, China
| | - Xin Guo
- Virus Laboratory, The Affiliated Shengjing Hospital, China Medical University, China
| | - Yaozhong Shao
- Virus Laboratory, The Affiliated Shengjing Hospital, China Medical University, China
| | - Zhengrong Sun
- Virus Laboratory, The Affiliated Shengjing Hospital, China Medical University, China
| | - Qiang Ruan
- Virus Laboratory, The Affiliated Shengjing Hospital, China Medical University, China.
| |
Collapse
|
18
|
Donohoe OH, Henshilwood K, Way K, Hakimjavadi R, Stone DM, Walls D. Identification and Characterization of Cyprinid Herpesvirus-3 (CyHV-3) Encoded MicroRNAs. PLoS One 2015; 10:e0125434. [PMID: 25928140 PMCID: PMC4416013 DOI: 10.1371/journal.pone.0125434] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/17/2015] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs involved in post-transcriptional gene regulation. Some viruses encode their own miRNAs and these are increasingly being recognized as important modulators of viral and host gene expression. Cyprinid herpesvirus 3 (CyHV-3) is a highly pathogenic agent that causes acute mass mortalities in carp (Cyprinus carpio carpio) and koi (Cyprinus carpio koi) worldwide. Here, bioinformatic analyses of the CyHV-3 genome suggested the presence of non-conserved precursor miRNA (pre-miRNA) genes. Deep sequencing of small RNA fractions prepared from in vitro CyHV-3 infections led to the identification of potential miRNAs and miRNA–offset RNAs (moRNAs) derived from some bioinformatically predicted pre-miRNAs. DNA microarray hybridization analysis, Northern blotting and stem-loop RT-qPCR were then used to definitively confirm that CyHV-3 expresses two pre-miRNAs during infection in vitro. The evidence also suggested the presence of an additional four high-probability and two putative viral pre-miRNAs. MiRNAs from the two confirmed pre-miRNAs were also detected in gill tissue from CyHV-3-infected carp. We also present evidence that one confirmed miRNA can regulate the expression of a putative CyHV-3-encoded dUTPase. Candidate homologues of some CyHV-3 pre-miRNAs were identified in CyHV-1 and CyHV-2. This is the first report of miRNA and moRNA genes encoded by members of the Alloherpesviridae family, a group distantly related to the Herpesviridae family. The discovery of these novel CyHV-3 genes may help further our understanding of the biology of this economically important virus and their encoded miRNAs may have potential as biomarkers for the diagnosis of latent CyHV-3.
Collapse
Affiliation(s)
- Owen H. Donohoe
- Marine Institute, Rinville, Oranmore, Co. Galway, Ireland
- School of Biotechnology and National Centre for Sensor Research, Dublin City University, Dublin, Ireland
| | | | - Keith Way
- Centre for Environment, Fisheries and Aquaculture Science (Cefas), The Nothe, Weymouth, Dorset, the United Kingdom
| | - Roya Hakimjavadi
- School of Biotechnology and National Centre for Sensor Research, Dublin City University, Dublin, Ireland
| | - David M. Stone
- Centre for Environment, Fisheries and Aquaculture Science (Cefas), The Nothe, Weymouth, Dorset, the United Kingdom
| | - Dermot Walls
- School of Biotechnology and National Centre for Sensor Research, Dublin City University, Dublin, Ireland
- * E-mail:
| |
Collapse
|
19
|
Abstract
The identification of virally encoded microRNAs (miRNAs) has had a major impact on the field of herpes virology. Given their ability to target cellular and viral transcripts, and the lack of immune response to small RNAs, miRNAs represent an ideal mechanism of gene regulation during viral latency and persistence. In this review, we discuss the role of miRNAs in virus latency and persistence, specifically focusing on herpesviruses. We cover the current knowledge on miRNAs in establishing and maintaining virus latency and promoting survival of infected cells through targeting of both viral and cellular transcripts, highlighting key publications in the field. We also discuss potential areas of future research and how novel technologies may aid in determining how miRNAs shape virus latency in the context of herpesvirus infections.
Collapse
Affiliation(s)
- Finn Grey
- Roslin Institute, Division of Infection and Immunity, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| |
Collapse
|
20
|
Human cytomegalovirus-encoded miR-US25-1 aggravates the oxidised low density lipoprotein-induced apoptosis of endothelial cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:531979. [PMID: 24895586 PMCID: PMC4033414 DOI: 10.1155/2014/531979] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/06/2014] [Accepted: 04/08/2014] [Indexed: 12/11/2022]
Abstract
Human cytomegalovirus (HCMV) infection is linked to the development and severity of the cardiovascular disease atherosclerosis; however, there is little known about the promotion of atherosclerosis. miR-US25-1 is one of HCMV-encoded miRNAs and targets cellular genes that are essential for virus growth to control the life cycle of the virus and host cells. The prominent regulation on cell cycle genes of the miR-US25-1 attracts us to explore its role in the atherosclerosis promotion. It was indicated that miR-US25-1 level was upregulated in subjects or in endothelial cells with HCMV infection; and the miR-US25-1 downregulated the expression of BRCC 3 by targeting the 5′ UTR of BRCC 3. And a miR-US25-1 mimics transfection could reduce the EAhy926 cell viability but did not induce apoptosis in EAhy926 cells. And what is more, miR-US25-1 mimicis transfection deteriorated the ox-LDL-induced apoptosis and aggravated the upregulation of apoptosis-associated molecules by oxidised low density lipoprotein (ox-LDL) in EAhy926 cells. And we have also confirmed the deregulation of BRCC 3 expression by miR-US25-1 by targeting the 5′ UTR of it. Given the vital role of BRCC 3 in DNA damage repairing, we speculated that the targeting inhibition of BRCC 3 by miR-US25-1 may contribute to the aggravation of ox-LDL-promoted apoptosis of endothelial EAhy926 cells.
Collapse
|
21
|
Hook L, Hancock M, Landais I, Grabski R, Britt W, Nelson JA. Cytomegalovirus microRNAs. Curr Opin Virol 2014; 7:40-6. [PMID: 24769092 DOI: 10.1016/j.coviro.2014.03.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 03/25/2014] [Accepted: 03/27/2014] [Indexed: 12/26/2022]
Abstract
The discovery that animals, plants and DNA viruses encode microRNAs (miRNAs) has transformed our understanding of the regulation of gene expression. miRNAs are ubiquitous small non-coding RNAs that regulate gene expression post-transcriptionally, generally by binding to sites within the 3' untranslated regions (UTR) of messenger RNA (mRNA) transcripts. To date, over 250 viral miRNAs have been identified primarily in members of the herpesvirus family. These viral miRNAs target both viral and cellular genes in order to regulate viral replication, the establishment and maintenance of viral latency, cell survival, and innate and adaptive immunity. This review will focus on our current knowledge of the targets and functions of human cytomegalovirus (HCMV) miRNAs and their functional equivalents in other herpesviruses.
Collapse
Affiliation(s)
- Lauren Hook
- VGTI, OHSU West Campus, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - Meaghan Hancock
- VGTI, OHSU West Campus, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - Igor Landais
- VGTI, OHSU West Campus, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - Robert Grabski
- Department of Pediatrics, University of Alabama, Birmingham, AL 35294, USA
| | - William Britt
- Department of Pediatrics, University of Alabama, Birmingham, AL 35294, USA
| | - Jay A Nelson
- VGTI, OHSU West Campus, 505 NW 185th Avenue, Beaverton, OR 97006, USA.
| |
Collapse
|
22
|
Analysis of viral microRNA expression by elephant endotheliotropic herpesvirus 1. Virology 2014; 454-455:102-8. [PMID: 24725936 DOI: 10.1016/j.virol.2014.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/14/2014] [Accepted: 02/09/2014] [Indexed: 11/21/2022]
Abstract
Elephant endotheliotropic herpesvirus 1 (EEHV1), a member of the Betaherpesvirinae subfamily, has recently emerged as an important viral pathogen of Asian elephants that can cause a severe, often fatal, hemorrhagic disease. EEHV1 does not replicate in culture and little is currently known about the molecular biology of this emerging pathogen, with the notable exception of its genomic DNA sequence. Here, we have used small RNA deep sequencing to determine whether EEHV1, like other human and murine betaherpesviruses, expresses viral microRNAs in infected tissues in vivo. Our data provide evidence supporting the existence of at least three novel viral microRNAs encoded by EEHV1 and one of these, miR-E3-5p, is shown to repress target mRNA expression. Moreover, miR-E3-5p expression was readily detectable in tissue samples derived from two infected elephants, including in whole blood. These data shed new light on the biology of EEHV1 and identify small RNAs that have the potential to be useful in the diagnosis of sub-clinical infections in captive Asian and African elephants.
Collapse
|
23
|
Parasite-derived microRNAs in host serum as novel biomarkers of helminth infection. PLoS Negl Trop Dis 2014; 8:e2701. [PMID: 24587461 PMCID: PMC3930507 DOI: 10.1371/journal.pntd.0002701] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 01/03/2014] [Indexed: 02/06/2023] Open
Abstract
Background MicroRNAs (miRNAs) are a class of short non-coding RNA that play important roles in disease processes in animals and are present in a highly stable cell-free form in body fluids. Here, we examine the capacity of host and parasite miRNAs to serve as tissue or serum biomarkers of Schistosoma mansoni infection. Methods/Principal Findings We used Exiqon miRNA microarrays to profile miRNA expression in the livers of mice infected with S. mansoni at 7 weeks post-infection. Thirty-three mouse miRNAs were differentially expressed in infected compared to naïve mice (>2 fold change, p<0.05) including miR-199a-3p, miR-199a-5p, miR-214 and miR-21, which have previously been associated with liver fibrosis in other settings. Five of the mouse miRNAs were also significantly elevated in serum by twelve weeks post-infection. Sequencing of small RNAs from serum confirmed the presence of these miRNAs and further revealed eleven parasite-derived miRNAs that were detectable by eight weeks post infection. Analysis of host and parasite miRNA abundance by qRT-PCR was extended to serum of patients from low and high infection sites in Zimbabwe and Uganda. The host-derived miRNAs failed to distinguish uninfected from infected individuals. However, analysis of three of the parasite-derived miRNAs (miR-277, miR-3479-3p and bantam) could detect infected individuals from low and high infection intensity sites with specificity/sensitivity values of 89%/80% and 80%/90%, respectively. Conclusions This work identifies parasite-derived miRNAs as novel markers of S. mansoni infection in both mice and humans, with the potential to be used with existing techniques to improve S. mansoni diagnosis. In contrast, although host miRNAs are differentially expressed in the liver during infection their abundance levels in serum are variable in human patients and may be useful in cases of extreme pathology but likely hold limited value for detecting prevalence of infection. Schistosomiasis is a chronic disease caused by blood flukes that affects over 200 million people worldwide, of which 90% live in Sub-Saharan Africa. In the field setting schistosomiasis caused by S. mansoni is diagnosed by detection of parasite eggs in stool samples using microscopic techniques. Here we investigate the potential of microRNAs (miRNAs), a class of short noncoding RNAs, to act as biomarkers of S. mansoni infection. We have identified a specific subset of murine miRNAs whose expression is significantly altered in the liver between 6–12 weeks post infection. However their abundance in serum is not significantly different between naïve and S. mansoni-infected mice until twelve weeks post infection and they do not display consistent differential abundance in the serum of infected versus uninfected humans. In contrast, three parasite-derived miRNAs (miR-277, bantam and miR-3479-3p) were detected in the serum of infected mice and human patients and the combined detection of these miRNAs could distinguish S. mansoni infected from uninfected individuals from low and high infection intensity areas with 89%/80% or 80%/90% specificity/sensitivity, respectively. These results demonstrate that miRNAs of parasite origin are a new class of serum biomarker for detecting S. mansoni and likely other helminth infections.
Collapse
|
24
|
Hook LM, Landais I, Hancock MH, Nelson JA. Techniques for characterizing cytomegalovirus-encoded miRNAs. Methods Mol Biol 2014; 1119:239-265. [PMID: 24639227 DOI: 10.1007/978-1-62703-788-4_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
microRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at the posttranscriptional level, by binding to sites within the 3' untranslated regions of messenger RNA (mRNA) transcripts. The discovery of this completely new mechanism of gene regulation necessitated the development of a variety of techniques to further characterize miRNAs, their expression, and function. In this chapter, we will discuss techniques currently used in the miRNA field to express, detect, and inhibit miRNAs as well as methods used to identify their targets.
Collapse
Affiliation(s)
- Lauren M Hook
- Vaccine & Gene Therapy Institute, Oregon Health and Sciences University, Beaverton, OR, USA
| | | | | | | |
Collapse
|
25
|
Schwarz TM, Volpe LAM, Abraham CG, Kulesza CA. Molecular investigation of the 7.2 kb RNA of murine cytomegalovirus. Virol J 2013; 10:348. [PMID: 24295514 PMCID: PMC4220806 DOI: 10.1186/1743-422x-10-348] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/22/2013] [Indexed: 11/10/2022] Open
Abstract
Background HCMV encodes a stable 5 kb RNA of unknown function that is conserved across cytomegalovirus species. In vivo studies of the MCMV orthologue, a 7.2 kb RNA, demonstrated that viruses that do not express the RNA fail to establish efficient persistent replication in the salivary glands of mice. To gain further insight into the function and properties of this conserved locus, we characterized the MCMV intron in finer detail. Methods We performed multiple analyses to evaluate transcript expression kinetics, identify transcript termini and promoter elements. The half-lives of intron locus RNAs were quantified by measuring RNA levels following actinomycin D treatment in a qRT-PCR-based assay. We also constructed a series of recombinant viruses to evaluate protein coding potential in the locus and test the role of putative promoter elements. These recombinant viruses were tested in both in vitro and in vivo assays. Results We show that the 7.2 kb RNA is expressed with late kinetics during productive infection of mouse fibroblasts. The termini of the precursor RNA that is processed to produce the intron were identified and we demonstrate that the m106 open reading frame, which resides on the spliced mRNA derived from precursor processing, can be translated during infection. Mapping the 5′ end of the primary transcript revealed minimal promoter elements located upstream that contribute to transcript expression. Analysis of recombinant viruses with deletions in the putative promoter elements, however, revealed these elements exert only minor effects on intron expression and viral persistence in vivo. Low transcriptional output by the putative promoter element(s) is compensated by the long half-life of the 7.2 kb RNA of approximately 28.8 hours. Detailed analysis of viral spread prior to the establishment of persistence also showed that the intron is not likely required for efficient spread to the salivary gland, but rather enhances persistent replication in this tissue site. Conclusions This data provides a comprehensive transcriptional analysis of the MCMV 7.2 kb intron locus. Our studies indicate that the 7.2 kb RNA is an extremely long-lived RNA, a feature which is likely to be important in its role promoting viral persistence in the salivary gland.
Collapse
Affiliation(s)
| | | | | | - Caroline A Kulesza
- Department of Microbiology, University of Colorado School of Medicine, MS8333, 12800 E, 19th Ave, Aurora, Colorado 80045, USA.
| |
Collapse
|
26
|
Functional genomics approaches to understand cytomegalovirus replication, latency and pathogenesis. Curr Opin Virol 2013; 3:408-15. [PMID: 23816389 DOI: 10.1016/j.coviro.2013.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 06/05/2013] [Accepted: 06/05/2013] [Indexed: 11/21/2022]
Abstract
Cytomegalovirus (CMV) is a species-specific herpesvirus that is ubiquitous in the population and has the potential to cause significant disease in immunocompromised individuals as well as in congenitally infected infants. CMV establishes latency in cells of the myeloid lineage following primary infection. High-throughput functional genomics approaches have provided insight into the mechanisms of CMV replication, but although CMV latency cell models have been useful in elucidating the mechanisms of viral latency and reactivation, omics approaches have proven challenging in these cell systems. This review will summarize the current state of knowledge concerning the use of functional genomics technologies to understand mechanisms of CMV replication, latency and pathogenesis.
Collapse
|
27
|
Abstract
Recently, it has become clear that herpesviruses are unique among pathogenic virus families in that they express multiple virally-encoded microRNAs in latently and/or lytically infected cells. The large size of herpesvirus genomes, combined with the inability of most human herpesviruses to replicate in animals, has until recently limited our ability to examine the contribution of viral miRNAs to herpesvirus replication and pathogenesis in vivo. However, recent data, primarily obtained using model animal herpesviruses, suggest that viral miRNAs, while not required for lytic replication in culture, can nevertheless strongly enhance viral pathogenesis, including oncogenesis, in vivo and also promote the establishment of a reservoir of latently infected cells.
Collapse
Affiliation(s)
- Bryan R Cullen
- Department of Molecular Genetics and Microbiology and Center for Virology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
28
|
Dittmer A, Förstemann K. Murine cytomegalovirus infection of cultured mouse cells induces expression of miR-7a. J Gen Virol 2012; 93:1537-1547. [PMID: 22442111 DOI: 10.1099/vir.0.041822-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
One goal of virus infection is to reprogramme the host cell to optimize virus replication. As part of this process, viral microRNAs (miRNAs) may compete for components of the miRNA/small interfering RNA pathway, as well as regulate cellular targets. Murine cytomegalovirus (MCMV) has been described to generate large numbers of viral miRNAs during lytic infection and was therefore used to analyse the impact of viral miRNAs on the host-cell small-RNA system, as well as to check for sorting of viral small RNAs into specific Argonaute (Ago) proteins. Deep-sequencing analysis of MCMV-infected cells revealed that viral miRNAs represented only ~13% of all detected miRNAs. All previously described MCMV miRNAs with the exception of miR-m88-1* were confirmed, and for the MCMV miR-m01-1 hairpin, an additional miRNA, designated miR-m01-1-3p, was found. Its presence was confirmed by quantitative real-time PCR and Northern blotting. Deep sequencing after RNA-induced silencing complex (RISC) immunoprecipitation with antibodies specific for either Ago1 or Ago2 showed that all MCMV miRNAs were loaded into both RISCs. The ratio of MCMV to mouse miRNAs was not increased after immunoprecipitation of Ago proteins. Viral miRNAs therefore did not overwhelm the host miRNA processing system, nor were they incorporated preferentially into RISCs. Three mouse miRNAs were found that showed altered expression as a result of MCMV infection. Downregulation of miR-27a, as described previously, could be confirmed. In addition, miR-26a was downregulated, and upregulation of miR-7a dependent on viral protein expression could be observed.
Collapse
Affiliation(s)
- Alexandra Dittmer
- Gene Center, Ludwig Maximilian University, Feodor Lynen Strasse 25, 81377 Munich, Germany
| | - Klaus Förstemann
- Gene Center, Ludwig Maximilian University, Feodor Lynen Strasse 25, 81377 Munich, Germany
| |
Collapse
|
29
|
Marcinowski L, Tanguy M, Krmpotic A, Rädle B, Lisnić VJ, Tuddenham L, Chane-Woon-Ming B, Ruzsics Z, Erhard F, Benkartek C, Babic M, Zimmer R, Trgovcich J, Koszinowski UH, Jonjic S, Pfeffer S, Dölken L. Degradation of cellular mir-27 by a novel, highly abundant viral transcript is important for efficient virus replication in vivo. PLoS Pathog 2012; 8:e1002510. [PMID: 22346748 PMCID: PMC3276556 DOI: 10.1371/journal.ppat.1002510] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 12/13/2011] [Indexed: 12/11/2022] Open
Abstract
Cytomegaloviruses express large amounts of viral miRNAs during lytic infection, yet, they only modestly alter the cellular miRNA profile. The most prominent alteration upon lytic murine cytomegalovirus (MCMV) infection is the rapid degradation of the cellular miR-27a and miR-27b. Here, we report that this regulation is mediated by the ∼1.7 kb spliced and highly abundant MCMV m169 transcript. Specificity to miR-27a/b is mediated by a single, apparently optimized, miRNA binding site located in its 3′-UTR. This site is easily and efficiently retargeted to other cellular and viral miRNAs by target site replacement. Expression of the 3′-UTR of m169 by an adenoviral vector was sufficient to mediate its function, indicating that no other viral factors are essential in this process. Degradation of miR-27a/b was found to be accompanied by 3′-tailing and -trimming. Despite its dramatic effect on miRNA stability, we found this interaction to be mutual, indicating potential regulation of m169 by miR-27a/b. Most interestingly, three mutant viruses no longer able to target miR-27a/b, either due to miRNA target site disruption or target site replacement, showed significant attenuation in multiple organs as early as 4 days post infection, indicating that degradation of miR-27a/b is important for efficient MCMV replication in vivo. MicroRNAs are small, non-coding RNAs which shape and fine-tune gene expression of at least a third of our genes. During millions of years of coevolution with their hosts, herpesviruses have both usurped the host cell miRNA machinery by expressing their own sets of miRNAs, and learned to modify host miRNA expression for their own needs. Recently, we reported on the rapid degradation of two cellular miRNAs upon lytic murine cytomegalovirus (MCMV) infection, namely miR-27a and miR-27b. In this paper, we show that their regulation is mediated by the highly abundant viral transcript m169. It targets miR-27a/b via a single binding site in its 3′-UTR, which can be efficiently retargeted to other cellular and viral miRNAs, enabling the efficient knock-down of individual miRNAs of interest. Degradation of miR-27a/b is preceded by its 3′-tailing and -trimming. Most interestingly, three mutant viruses unable to target miR-27a/b showed significantly lower virus titers in various organs during acute MCMV infection, indicating that degradation of miR-27a/b is important for efficient virus replication in vivo.
Collapse
Affiliation(s)
- Lisa Marcinowski
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Mélanie Tanguy
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
| | - Astrid Krmpotic
- Department of Histology and Embryology, Faculty of Medicine University of Rijeka, Rijeka, Croatia
| | - Bernd Rädle
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Vanda J. Lisnić
- Department of Histology and Embryology, Faculty of Medicine University of Rijeka, Rijeka, Croatia
| | - Lee Tuddenham
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
| | - Béatrice Chane-Woon-Ming
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
| | - Zsolt Ruzsics
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Florian Erhard
- Institute for Informatics, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Marina Babic
- Department of Histology and Embryology, Faculty of Medicine University of Rijeka, Rijeka, Croatia
| | - Ralf Zimmer
- Institute for Informatics, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Joanne Trgovcich
- Department of Pathology, The Ohio State University, Columbus, Ohio, United States of America
| | - Ulrich H. Koszinowski
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Stipan Jonjic
- Department of Histology and Embryology, Faculty of Medicine University of Rijeka, Rijeka, Croatia
- * E-mail: (SJ); (SP); (LD)
| | - Sébastien Pfeffer
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
- * E-mail: (SJ); (SP); (LD)
| | - Lars Dölken
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
- * E-mail: (SJ); (SP); (LD)
| |
Collapse
|
30
|
Rhesus cytomegalovirus encodes seventeen microRNAs that are differentially expressed in vitro and in vivo. Virology 2012; 425:133-42. [PMID: 22305624 DOI: 10.1016/j.virol.2012.01.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 11/29/2011] [Accepted: 01/11/2012] [Indexed: 02/07/2023]
Abstract
Human cytomegalovirus (HCMV) miRNAs are important for regulation of viral infection and evasion of host immune responses. Unfortunately, the importance of HCMV miRNAs cannot be addressed in vivo due to the species specificity of CMVs. Rhesus CMV (RhCMV) infection of rhesus macaques provides an important model system for HCMV pathogenesis due to the genetic similarity between the viruses. In this report, seventeen RhCMV miRNAs were identified using Next Generation Sequencing. In fibroblasts, RhCMV miRNAs associate with Argonaute proteins and display several patterns of expression, including an early peak in expression followed by decline and accumulation throughout infection. Additionally, RhCMV encodes an HCMV miR-US5-2 homologue that targets the 3' UTR of RhCMV US7. Finally, examination of salivary gland tissue from infected animals revealed the presence of a subset of viral miRNAs. This study highlights the importance of the RhCMV model system for evaluating the roles of CMV miRNAs during viral infection.
Collapse
|
31
|
Tuddenham L, Jung JS, Chane-Woon-Ming B, Dölken L, Pfeffer S. Small RNA deep sequencing identifies microRNAs and other small noncoding RNAs from human herpesvirus 6B. J Virol 2012; 86:1638-49. [PMID: 22114334 PMCID: PMC3264354 DOI: 10.1128/jvi.05911-11] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 11/11/2011] [Indexed: 02/07/2023] Open
Abstract
Roseolovirus, or human herpesvirus 6 (HHV-6), is a ubiquitous human pathogen infecting over 95% of the population by the age of 2 years. As with other herpesviruses, reactivation of HHV-6 can present with severe complications in immunocompromised individuals. Recent studies have highlighted the importance of herpesvirus-derived microRNAs (miRNAs) in modulating both cellular and viral gene expression. An initial report which computed the likelihood of various viruses to encode miRNAs did not predict HHV-6 miRNAs. To experimentally screen for small HHV-6-encoded RNAs, we conducted large-scale sequencing of Sup-T-1 cells lytically infected with a laboratory strain of HHV-6B. This revealed an abundant, 60- to 65-nucleotide RNA of unknown function derived from the lytic origin of replication (OriLyt) that gave rise to smaller RNA species of 18 or 19 nucleotides. In addition, we identified four pre-miRNAs whose mature forms accumulated in Argonaute 2. In contrast to the case for other betaherpesviruses, HHV-6B miRNAs are expressed from direct repeat regions (DR(L) and DR(R)) located at either side of the genome. All miRNAs are conserved in the closely related HHV-6A variant, and one of them is a seed ortholog of the human miRNA miR-582-5p. Similar to alphaherpesvirus miRNAs, they are expressed in antisense orientation relative to immediate-early open reading frames (ORFs) and thus have the potential to regulate key viral genes.
Collapse
Affiliation(s)
- Lee Tuddenham
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
| | - Jette S. Jung
- Max von Pettenkofer Institute, Ludwig Maximilians University Munich, Munich, Germany
| | - Béatrice Chane-Woon-Ming
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
| | - Lars Dölken
- Max von Pettenkofer Institute, Ludwig Maximilians University Munich, Munich, Germany
- University of Cambridge, Department of Medicine, Cambridge, United Kingdom
| | - Sébastien Pfeffer
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
| |
Collapse
|
32
|
Libri V, Helwak A, Miesen P, Santhakumar D, Borger JG, Kudla G, Grey F, Tollervey D, Buck AH. Murine cytomegalovirus encodes a miR-27 inhibitor disguised as a target. Proc Natl Acad Sci U S A 2012; 109:279-84. [PMID: 22184245 PMCID: PMC3252920 DOI: 10.1073/pnas.1114204109] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Individual microRNAs (miRNAs) are rapidly down-regulated during conditions of cellular activation and infection, but factors mediating miRNA turnover are poorly understood. Infection of mouse cells with murine cytomegalovirus (MCMV) induces the rapid down-regulation of an antiviral cellular miRNA, miR-27. Here, we identify a transcript produced by MCMV that binds to miR-27 and mediates its degradation. UV-crosslinking and high-throughput sequencing [CRAC (UV-crosslinking and analysis of cDNA)] identified MCMV RNA segments associated with the miRNA-binding protein Argonaute 2 (Ago2). A cluster of hits mapped to a predicted miR-27-binding site in the 3'UTR of the previously uncharacterized ORF, m169. The expression kinetics of the m169 transcript correlated with degradation of miR-27 during infection, and m169 expression inhibited miR-27 functional activity in a reporter assay. siRNA knockdown of m169 demonstrated its requirement for miR-27 degradation following infection and did not affect other host miRNAs. Substitution of the miR-27-binding site in m169 to create complementarity to a different cellular miRNA, miR-24, resulted in down-regulation of only miR-24 following infection. The m169 transcript is cytoplasmic, capped, polyadenylated, and interacts with miRNA-27 through seed pairing: characteristic features of the normal messenger RNA (mRNA) targets of miRNAs. This virus-host interaction reveals a mode of miRNA regulation in which a mRNA directs the degradation of a miRNA. We speculate that RNA-mediated miRNA degradation could be a more general viral strategy for manipulating host cells.
Collapse
Affiliation(s)
- Valentina Libri
- Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| | - Aleksandra Helwak
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| | - Pascal Miesen
- Department of Medical Microbiology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, 6500 HB, Nijmegen, The Netherlands
| | - Diwakar Santhakumar
- Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
- Division of Pathway Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; and
| | - Jessica G. Borger
- Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| | - Grzegorz Kudla
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| | - Finn Grey
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, United Kingdom
| | - David Tollervey
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| | - Amy H. Buck
- Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
- Division of Pathway Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; and
| |
Collapse
|
33
|
Oulas A, Karathanasis N, Louloupi A, Poirazi P. Finding cancer-associated miRNAs: methods and tools. Mol Biotechnol 2011; 49:97-107. [PMID: 21607762 DOI: 10.1007/s12033-011-9416-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Changes in the structure and/or the expression of protein coding genes were thought to be the major cause of cancer for many decades. The recent discovery of non-coding RNA (ncRNA) transcripts (i.e., microRNAs) suggests that the molecular biology of cancer is far more complex. MicroRNAs (miRNAs) have been under investigation due to their involvement in carcinogenesis, often taking up roles of tumor suppressors or oncogenes. Due to the slow nature of experimental identification of miRNA genes, computational procedures have been applied as a valuable complement to cloning. Numerous computational tools, implemented to recognize the features of miRNA biogenesis, have resulted in the prediction of novel miRNA genes. Computational approaches provide clues as to which are the dominant features that characterize these regulatory units and furthermore act by narrowing down the search space making experimental verification faster and cheaper. In combination with large scale, high throughput methods, such as deep sequencing, computational methods have aided in the discovery of putative molecular signatures of miRNA deregulation in human tumors. This review focuses on existing computational methods for identifying miRNA genes, provides an overview of the methodology undertaken by these tools, and underlies their contribution towards unraveling the role of miRNAs in cancer.
Collapse
Affiliation(s)
- Anastasis Oulas
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Crete, Greece
| | | | | | | |
Collapse
|
34
|
Tuddenham L, Pfeffer S. Roles and regulation of microRNAs in cytomegalovirus infection. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2011; 1809:613-22. [DOI: 10.1016/j.bbagrm.2011.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 04/12/2011] [Accepted: 04/14/2011] [Indexed: 12/21/2022]
|
35
|
Cullen BR. Viruses and microRNAs: RISCy interactions with serious consequences. Genes Dev 2011; 25:1881-94. [PMID: 21896651 DOI: 10.1101/gad.17352611] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Analyses of small RNA expression profiles have revealed that several DNA viruses-including particularly, herpesviruses-express high levels of multiple viral microRNAs (miRNAs) in infected cells. Here, I review our current understanding of how viral miRNAs influence viral replication and pathogenesis and discuss how viruses reshape the pattern of cellular miRNA expression. Indeed, viruses are now known to both activate and repress the expression of specific cellular miRNAs, and disrupting this process can perturb the ability of viruses to replicate normally. In addition, it is now clear that virally encoded miRNAs play a key role in inhibiting antiviral innate immune responses and can also promote cell transformation in culture. While our understanding of how viruses interact with miRNAs remains somewhat rudimentary, it is nevertheless already clear that these interactions can play a critical role in mediating viral pathogenesis and therefore may represent novel and highly specific targets for therapeutic intervention.
Collapse
Affiliation(s)
- Bryan R Cullen
- Department of Molecular Genetics and Microbiology, Center for Virology, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
36
|
All is fair in virus-host interactions: NK cells and cytomegalovirus. Trends Mol Med 2011; 17:677-85. [PMID: 21852192 DOI: 10.1016/j.molmed.2011.07.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 07/11/2011] [Accepted: 07/12/2011] [Indexed: 12/12/2022]
Abstract
The infection of mice with mouse cytomegalovirus (MCMV) as a model of human cytomegalovirus (HCMV) infection has been particularly informative in elucidating the role of innate and adaptive immune response mechanisms during infection. Millions of years of co-evolution between cytomegaloviruses (CMV) and their hosts has resulted in numerous attempts to overwhelm each other. CMVs devote many genes to modulating the host natural killer (NK) cell response and NK cells employ many strategies to cope with CMV infection. While focusing on these attack-counterattack measures, this review will discuss several novel mechanisms of immune evasion by MCMV, the role of Ly49 receptors in mediating resistance to MCMV, and the impact of the initial NK cell response on the shaping of adaptive immunity.
Collapse
|
37
|
Jurak I, Griffiths A, Coen DM. Mammalian alphaherpesvirus miRNAs. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2011; 1809:641-53. [PMID: 21736960 DOI: 10.1016/j.bbagrm.2011.06.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 06/17/2011] [Accepted: 06/18/2011] [Indexed: 12/26/2022]
Abstract
Mammalian alphaherpesviruses are major causes of human and veterinary disease. During productive infection, these viruses exhibit complex and robust patterns of gene expression. These viruses also form latent infections in neurons of sensory ganglia in which productive cycle gene expression is highly repressed. Both modes of infection provide advantageous opportunities for regulation by microRNAs. Thus far, published data regarding microRNAs are available for six mammalian alphaherpesviruses. No microRNAs have yet been detected from varicella zoster virus. The five other viruses-herpes simplex viruses-1 and -2, herpes B virus, bovine herpesvirus-1, and pseudorabies virus-representing both genera of mammalian alphaherpesviruses have been shown to express microRNAs. In this article, we discuss these microRNAs in terms of where they are encoded in the viral genome relative to other viral transcripts; whether they are expressed during productive or latent infection; their potential targets; what little is known about their actual targets and functions during viral infection; and what little is known about the interactions of these viruses with the host microRNA machinery. This article is part of a Special Issue entitled: "MicroRNAs in viral gene regulation".
Collapse
Affiliation(s)
- Igor Jurak
- Department of Biological Chemistry, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
38
|
Lacaze P, Forster T, Ross A, Kerr LE, Salvo-Chirnside E, Lisnic VJ, López-Campos GH, García-Ramírez JJ, Messerle M, Trgovcich J, Angulo A, Ghazal P. Temporal profiling of the coding and noncoding murine cytomegalovirus transcriptomes. J Virol 2011; 85:6065-76. [PMID: 21471238 PMCID: PMC3126304 DOI: 10.1128/jvi.02341-10] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 03/28/2011] [Indexed: 12/20/2022] Open
Abstract
The global transcriptional program of murine cytomegalovirus (MCMV), involving coding, noncoding, and antisense transcription, remains unknown. Here we report an oligonucleotide custom microarray platform capable of measuring both coding and noncoding transcription on a genome-wide scale. By profiling MCMV wild-type and immediate-early mutant strains in fibroblasts, we found rapid activation of the transcriptome by 6.5 h postinfection, with absolute dependency on ie3, but not ie1 or ie2, for genomic programming of viral gene expression. Evidence is also presented to show, for the first time, genome-wide noncoding and bidirectional transcription at late stages of MCMV infection.
Collapse
Affiliation(s)
- Paul Lacaze
- Division of Pathway Medicine, The University of Edinburgh, The Chancellor's Building, College of Medicine, 49 Little France Crescent, Edinburgh, United Kingdom
| | - Thorsten Forster
- Division of Pathway Medicine, The University of Edinburgh, The Chancellor's Building, College of Medicine, 49 Little France Crescent, Edinburgh, United Kingdom
| | - Alan Ross
- Division of Pathway Medicine, The University of Edinburgh, The Chancellor's Building, College of Medicine, 49 Little France Crescent, Edinburgh, United Kingdom
| | - Lorraine E. Kerr
- Centre for Systems Biology at Edinburgh, The University of Edinburgh, Darwin Building, King's Buildings Campus, Mayfield Road, Edinburgh, United Kingdom
| | - Eliane Salvo-Chirnside
- Centre for Systems Biology at Edinburgh, The University of Edinburgh, Darwin Building, King's Buildings Campus, Mayfield Road, Edinburgh, United Kingdom
| | - Vanda Juranic Lisnic
- Department of Histology and Embryology, Faculty of Medicine, Rijeka University, Croatia
| | | | - José J. García-Ramírez
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla—La Mancha, Avenida de Almansa 14, 02006 Albacete, Spain
| | - Martin Messerle
- Department of Virology, Hannover Medical School, Hannover, Germany
| | - Joanne Trgovcich
- Department of Pathology, The Ohio State University, Columbus, Ohio 43210
| | - Ana Angulo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Peter Ghazal
- Division of Pathway Medicine, The University of Edinburgh, The Chancellor's Building, College of Medicine, 49 Little France Crescent, Edinburgh, United Kingdom
- Centre for Systems Biology at Edinburgh, The University of Edinburgh, Darwin Building, King's Buildings Campus, Mayfield Road, Edinburgh, United Kingdom
| |
Collapse
|
39
|
Steitz J, Borah S, Cazalla D, Fok V, Lytle R, Mitton-Fry R, Riley K, Samji T. Noncoding RNPs of viral origin. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a005165. [PMID: 20719877 DOI: 10.1101/cshperspect.a005165] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Like their host cells, many viruses produce noncoding (nc)RNAs. These show diversity with respect to time of expression during viral infection, length and structure, protein-binding partners and relative abundance compared with their host-cell counterparts. Viruses, with their limited genomic capacity, presumably evolve or acquire ncRNAs only if they selectively enhance the viral life cycle or assist the virus in combating the host's response to infection. Despite much effort, identifying the functions of viral ncRNAs has been extremely challenging. Recent technical advances and enhanced understanding of host-cell ncRNAs promise accelerated insights into the RNA warfare mounted by this fascinating class of RNPs.
Collapse
Affiliation(s)
- Joan Steitz
- Department of Molecular Biophysics and Biochemistry, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06536-0812, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Grundhoff A, Sullivan CS. Virus-encoded microRNAs. Virology 2011; 411:325-43. [PMID: 21277611 DOI: 10.1016/j.virol.2011.01.002] [Citation(s) in RCA: 323] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Accepted: 01/04/2011] [Indexed: 01/03/2023]
Abstract
MicroRNAs (miRNAs) are the subject of enormous interest. They are small non-coding RNAs that play a regulatory role in numerous and diverse cellular processes such as immune function, apoptosis and tumorigenesis. Several virus families have been shown to encode miRNAs, and an appreciation for their roles in the viral infectious cycle continues to grow. Despite the identification of numerous (>225) viral miRNAs, an in depth functional understanding of most virus-encoded miRNAs is lacking. Here we focus on a few viral miRNAs with well-defined functions. We use these examples to extrapolate general themes of viral miRNA activities including autoregulation of viral gene expression, avoidance of host defenses, and a likely important role in maintaining latent and persistent infections. We hypothesize that although the molecular mechanisms and machinery are similar, the majority of viral miRNAs may utilize a target strategy that differs from host miRNAs. That is, many viral miRNAs may have evolved to regulate viral-encoded transcripts or networks of host genes that are unique to viral miRNAs. Included in this latter category is a likely abundant class of viral miRNAs that may regulate only one or a few principal host genes. Key steps forward for the field are discussed, including the need for additional functional studies that utilize surgical viral miRNA mutants combined with relevant models of infection.
Collapse
Affiliation(s)
- Adam Grundhoff
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Martinistr, Hamburg, Germany.
| | | |
Collapse
|
41
|
Abstract
Changes in the structure and/or the expression of protein-coding genes were thought to be the major cause of cancer for many decades. However, the recent discovery of non-coding RNA (ncRNA) transcripts suggests that the molecular biology of cancer is far more complex. MicroRNAs (miRNAs) are key players of the family of ncRNAs and they have been under extensive investigation because of their involvement in carcinogenesis, often taking up roles of tumor suppressors or oncogenes. Owing to the slow nature of experimental identification of miRNA genes, computational procedures have been applied as a valuable complement to cloning. Numerous computational tools, implemented to recognize the characteristic features of miRNA biogenesis, have resulted in the prediction of multiple novel miRNA genes. Computational approaches provide valuable clues as to which are the dominant features that characterize these regulatory units and furthermore act by narrowing down the search space making experimental verification faster and significantly cheaper. Moreover, in combination with large-scale, high-throughput methods, such as deep sequencing and tilling arrays, computational methods have aided in the discovery of putative molecular signatures of miRNA deregulation in human tumors. This chapter focuses on existing computational methods for identifying miRNA genes, provides an overview of the methodology undertaken by these tools, and underlies their contribution toward unraveling the role of miRNAs in cancer.
Collapse
Affiliation(s)
- Anastasis Oulas
- Institute for Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Crete, Greece
| | | | | |
Collapse
|
42
|
Abstract
One of the most significant recent advances in biomedical research has been the discovery of the approximately 22-nt-long class of noncoding RNAs designated microRNAs (miRNAs). These regulatory RNAs provide a unique level of posttranscriptional gene regulation that modulates a range of fundamental cellular processes. Several viruses, especially herpesviruses, also encode miRNAs, and over 200 viral miRNAs have now been identified. Current evidence indicates that viruses use these miRNAs to manipulate both cellular and viral gene expression. Furthermore, viral infection can exert a profound impact on the cellular miRNA expression profile, and several RNA viruses have been reported to interact directly with cellular miRNAs and/or to use these miRNAs to augment their replication potential. Here we discuss our current knowledge of viral miRNAs and virally influenced cellular miRNAs and their relationship to viral infection.
Collapse
Affiliation(s)
- Rebecca L Skalsky
- Department of Molecular Genetics and Microbiology and Center for Virology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | |
Collapse
|
43
|
Abstract
One of the most significant recent advances in biomedical research has been the discovery of the approximately 22-nt-long class of noncoding RNAs designated microRNAs (miRNAs). These regulatory RNAs provide a unique level of posttranscriptional gene regulation that modulates a range of fundamental cellular processes. Several viruses, especially herpesviruses, also encode miRNAs, and over 200 viral miRNAs have now been identified. Current evidence indicates that viruses use these miRNAs to manipulate both cellular and viral gene expression. Furthermore, viral infection can exert a profound impact on the cellular miRNA expression profile, and several RNA viruses have been reported to interact directly with cellular miRNAs and/or to use these miRNAs to augment their replication potential. Here we discuss our current knowledge of viral miRNAs and virally influenced cellular miRNAs and their relationship to viral infection.
Collapse
Affiliation(s)
- Rebecca L Skalsky
- Department of Molecular Genetics and Microbiology and Center for Virology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | |
Collapse
|
44
|
Cytomegalovirus microRNA expression is tissue specific and is associated with persistence. J Virol 2010; 85:378-89. [PMID: 20980502 DOI: 10.1128/jvi.01900-10] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small noncoding RNAs involved in posttranscriptional regulation. miRNAs are utilized in organisms ranging from plants to higher mammals, and data have shown that DNA viruses also use this method for host and viral gene regulation. Here, we report the sequencing of the small RNAs in rat cytomegalovirus (RCMV)-infected fibroblasts and persistently infected salivary glands. We identified 24 unique miRNAs that mapped to hairpin structures found within the viral genome. While most miRNAs were detected in both samples, four were detected exclusively in the infected fibroblasts and two were specific for the infected salivary glands. The RCMV miRNAs are distributed across the viral genome on both the positive and negative strands, with clusters of miRNAs at a number of locations, including near viral genes r1 and r111. The RCMV miRNAs have a genomic positional orientation similar to that of the miRNAs described for mouse cytomegalovirus, but they do not share any substantial sequence conservation. Similar to other reported miRNAs, the RCMV miRNAs had considerable variation at their 3' and 5' ends. Interestingly, we found a number of specific examples of differential isoform usage between the fibroblast and salivary gland samples. We determined by real-time PCR that expression of the RCMV miRNA miR-r111.1-2 is highly expressed in the salivary glands and that miR-R87-1 is expressed in most tissues during the acute infection phase. Our study identified the miRNAs expressed by RCMV in vitro and in vivo and demonstrated that expression is tissue specific and associated with a stage of viral infection.
Collapse
|
45
|
Dhuruvasan K, Sivasubramanian G, Pellett PE. Roles of host and viral microRNAs in human cytomegalovirus biology. Virus Res 2010; 157:180-92. [PMID: 20969901 DOI: 10.1016/j.virusres.2010.10.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 10/11/2010] [Accepted: 10/12/2010] [Indexed: 02/02/2023]
Abstract
Human cytomegalovirus (HCMV) has a relatively large and complex genome, a protracted lytic replication cycle, and employs a strategy of replicational latency as part of its lifelong persistence in the infected host. An important form of gene regulation in plants and animals revolves around a type of small RNA known as microRNA (miRNA). miRNAs can serve as major regulators of key developmental pathways, as well as provide subtle forms of regulatory control. The human genome encodes over 900 miRNAs, and miRNAs are also encoded by some viruses, including HCMV, which encodes at least 14 miRNAs. Some of the HCMV miRNAs are known to target both viral and cellular genes, including important immunomodulators. In addition to expressing their own miRNAs, infections with some viruses, including HCMV, can result in changes in the expression of cellular miRNAs that benefit virus replication. In this review, we summarize the connections between miRNAs and HCMV biology. We describe the nature of miRNA genes, miRNA biogenesis and modes of action, methods for studying miRNAs, HCMV-encoded miRNAs, effects of HCMV infection on cellular miRNA expression, roles of miRNAs in HCMV biology, and possible HCMV-related diagnostic and therapeutic applications of miRNAs.
Collapse
Affiliation(s)
- Kavitha Dhuruvasan
- Department of Immunology and Microbiology, Wayne State University School of Medicine, 540 East Canfield Avenue, 6225 Scott Hall, Detroit, MI 48201, USA.
| | | | | |
Collapse
|
46
|
Dölken L, Krmpotic A, Kothe S, Tuddenham L, Tanguy M, Marcinowski L, Ruzsics Z, Elefant N, Altuvia Y, Margalit H, Koszinowski UH, Jonjic S, Pfeffer S. Cytomegalovirus microRNAs facilitate persistent virus infection in salivary glands. PLoS Pathog 2010; 6:e1001150. [PMID: 20976200 PMCID: PMC2954898 DOI: 10.1371/journal.ppat.1001150] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Accepted: 09/10/2010] [Indexed: 12/22/2022] Open
Abstract
Micro (mi)RNAs are small non-coding RNAs that regulate the expression of their targets' messenger RNAs through both translational inhibition and regulation of target RNA stability. Recently, a number of viruses, particularly of the herpesvirus family, have been shown to express their own miRNAs to control both viral and cellular transcripts. Although some targets of viral miRNAs are known, their function in a physiologically relevant infection remains to be elucidated. As such, no in vivo phenotype of a viral miRNA knock-out mutant has been described so far. Here, we report on the first functional phenotype of a miRNA knock-out virus in vivo. During subacute infection of a mutant mouse cytomegalovirus lacking two viral miRNAs, virus production is selectively reduced in salivary glands, an organ essential for virus persistence and horizontal transmission. This phenotype depends on several parameters including viral load and mouse genetic background, and is abolished by combined but not single depletion of natural killer (NK) and CD4+ T cells. Together, our results point towards a miRNA-based immunoevasion mechanism important for long-term virus persistence.
Collapse
Affiliation(s)
- Lars Dölken
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Astrid Krmpotic
- Department of Histology and Embryology, Faculty of Medicine University of Rijeka, Rijeka, Croatia
| | - Sheila Kothe
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lee Tuddenham
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
| | - Mélanie Tanguy
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
| | - Lisa Marcinowski
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Zsolt Ruzsics
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Naama Elefant
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Yael Altuvia
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Hanah Margalit
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Ulrich H. Koszinowski
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Stipan Jonjic
- Department of Histology and Embryology, Faculty of Medicine University of Rijeka, Rijeka, Croatia
- * E-mail: (SJ); (SP)
| | - Sébastien Pfeffer
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire du CNRS, Strasbourg, France
- * E-mail: (SJ); (SP)
| |
Collapse
|
47
|
Abstract
In eukaryotic RNA-based antiviral immunity, viral double-stranded RNA is recognized as a pathogen-associated molecular pattern and processed into small interfering RNAs (siRNAs) by the host ribonuclease Dicer. After amplification by host RNA-dependent RNA polymerases in some cases, these virus-derived siRNAs guide specific antiviral immunity through RNA interference and related RNA silencing effector mechanisms. Here, I review recent studies on the features of viral siRNAs and other virus-derived small RNAs from virus-infected fungi, plants, insects, nematodes and vertebrates and discuss the innate and adaptive properties of RNA-based antiviral immunity.
Collapse
Affiliation(s)
- Shou-Wei Ding
- Department of Plant Pathology and Microbiology, and Institute for Integrative Genome Biology, University of California, Riverside, California 92521, USA.
| |
Collapse
|
48
|
Lin YT, Kincaid RP, Arasappan D, Dowd SE, Hunicke-Smith SP, Sullivan CS. Small RNA profiling reveals antisense transcription throughout the KSHV genome and novel small RNAs. RNA (NEW YORK, N.Y.) 2010; 16:1540-1558. [PMID: 20566670 PMCID: PMC2905754 DOI: 10.1261/rna.1967910] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Accepted: 04/27/2010] [Indexed: 05/29/2023]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is a human tumor virus that encodes 12 precursor microRNAs (pre-miRNAs) that give rise to 17 different known approximately 22-nucleotide (nt) effector miRNAs. Like all herpesviruses, KSHV has two modes of infection: (1) a latent mode whereby only a subset of viral genes are expressed and (2) a lytic mode during which the full remaining viral genes are expressed. To date, KSHV miRNAs have been mostly identified via analysis of cells that are undergoing latent infection. Here, we developed a method to profile small RNAs ( approximately 18-75 nt) from populations of cells undergoing predominantly lytic infection. Using two different next-generation sequencing platforms, we cloned and sequenced both pre-miRNAs and derivative miRNAs. Our analysis shows that the vast majority of viral and host 5p miRNAs are co-terminal with the 5' end of the cloned pre-miRNAs, consistent with both being defined by microprocessor cleavage. We report the complete repertoire (25 total) of 5p and 3p derivative miRNAs from all 12 previously described KSHV pre-miRNAs. Two KSHV pre-miRNAs, pre-miR-K12-8 and pre-miR-K12-12, encode abundant derivative miRNAs from the previously unreported strands of the pre-miRNA. We identify several novel small RNAs of low abundance, including viral miRNA-offset-RNAs (moRNAs), and antisense viral miRNAs (miRNA-AS) that are encoded antisense to previously reported KSHV pre-miRNAs. Finally, we observe widespread antisense transcription relative to known coding sequences during lytic replication. Despite the enormous potential to form double-stranded RNA in KSHV-infected cells, we observe no evidence for the existence of abundant viral-derived small interfering RNAs (siRNAs).
Collapse
Affiliation(s)
- Yao-Tang Lin
- Molecular Genetics and Microbiology, The University of Texas at Austin, Austin, TX 78712-0162, USA
| | | | | | | | | | | |
Collapse
|
49
|
Mutations in the M112/M113-coding region facilitate murine cytomegalovirus replication in human cells. J Virol 2010; 84:7994-8006. [PMID: 20519391 DOI: 10.1128/jvi.02624-09] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cytomegaloviruses, representatives of the Betaherpesvirinae, cause opportunistic infections in immunocompromised hosts. They infect various cells and tissues in their natural host but are highly species specific. For instance, human cytomegalovirus (HCMV) does not replicate in mouse cells, and human cells are not permissive for murine cytomegalovirus (MCMV) infection. However, the underlying molecular mechanisms are so far poorly understood. In the present study we isolated and characterized a spontaneously occurring MCMV mutant that has gained the capacity to replicate rapidly and to high titers in human cells. Compared to the parental wild-type (wt) virus, this mutant formed larger nuclear replication compartments and replicated viral DNA more efficiently. It also disrupted promyelocytic leukemia (PML) protein nuclear domains with greater efficiency but caused less apoptosis than did wt MCMV. Sequence analysis of the mutant virus genome revealed mutations in the M112/M113-coding region. This region is homologous to the HCMV UL112-113 region and encodes the viral early 1 (E1) proteins, which are known to play an important role in viral DNA replication. By introducing the M112/M113 mutations into wt MCMV, we demonstrated that they are sufficient to facilitate MCMV replication in human cells and are, at least in part, responsible for the efficient replication capability of the spontaneously adapted virus. However, additional mutations probably contribute as well. These results reveal a previously unrecognized role of the viral E1 proteins in regulating viral replication in different cells and provide new insights into the mechanisms of the species specificity of cytomegaloviruses.
Collapse
|
50
|
Chang Y, Jiang HJ, Sun XM, Cai XK, He XX, Li PY, Tang WX, Song YH, Lin JS. Hepatic stellate cell-specific gene silencing induced by an artificial microRNA for antifibrosis in vitro. Dig Dis Sci 2010; 55:642-53. [PMID: 19890714 DOI: 10.1007/s10620-009-1021-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Accepted: 09/30/2009] [Indexed: 12/14/2022]
Abstract
BACKGROUND We previously reported that the anti-transforming growth factor-beta1 (TGF-beta1) ribozymes directed by T7 and CMV promoters could reverse the character of activated hepatic stellate cells (HSCs) in vitro and improve fibrotic pathology in vivo. However, nonspecific elimination of the effects of TGF-beta1 without selectivity might have unfavorable consequences, such as overwhelming inflammation, tissue necrosis, etc. AIMS To establish an activated-HSC-specific gene silencing method and validate its feasibility for antifibrosis in vitro. METHODS An artificial intronic microRNA (miRNA) expression system was established, containing three parts: (1) a 1,074-bp SM-alpha actin promoter SMP8, which is a kind of RNA polymerase II promoter and has no activity in normal liver-derived cells but is switched on during the activation of HSCs, (2) intron1 modified by inserting an artificial pre-miRNA sequence against TGF-beta1, and (3) report gene enhanced green fluorescent proteins (EGFP). The feasibility of this system for artificial microRNA expression was validated through microRNA detection by real-time polymerase chain reaction (PCR). Alteration of biological characteristics of HSCs with the anti-TGF-beta1 miRNAs was preliminarily evaluated by measuring the expression levels of TGF-beta1 and its downstream molecules, including collagen I, matrix metalloproteinase 2 (MMP2), tissue inhibitor of metalloproteinase 1 (TIMP-1), etc. RESULTS The microRNA expression system could successfully produce mature anti-TGF-beta1 miRNAs in an activated-HSC-specific manner. The microRNA-induced inhibition rate of TGF-beta1 reached 70% and above. Accompanied by TGF-beta1 suppression, its downstream targets such as collagen I, MMP2, TIMP-1, etc. were also significantly downregulated in vitro. CONCLUSIONS Activated-HSC-cell-specific gene silencing could be induced well by the artificial intronic microRNA expression system to realize antifibrosis in vitro.
Collapse
Affiliation(s)
- Ying Chang
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | | | | | | | | | |
Collapse
|