151
|
Mouse cytomegalovirus microRNAs dominate the cellular small RNA profile during lytic infection and show features of posttranscriptional regulation. J Virol 2007; 81:13771-82. [PMID: 17942535 DOI: 10.1128/jvi.01313-07] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
MicroRNAs (miRNAs) are small, noncoding RNA molecules that regulate gene expression at the posttranscriptional level. Originally identified in a variety of organisms ranging from plants to mammals, miRNAs have recently been identified in several viruses. Viral miRNAs may play a role in modulating both viral and host gene expression. Here, we report on the identification and characterization of 18 viral miRNAs from mouse fibroblasts lytically infected with the murine cytomegalovirus (MCMV). The MCMV miRNAs are expressed at early times of infection and are scattered in small clusters throughout the genome with up to four distinct miRNAs processed from a single transcript. No significant homologies to human CMV-encoded miRNAs were found. Remarkably, as soon as 24 h after infection, MCMV miRNAs constituted about 35% of the total miRNA pool, and at 72 h postinfection, this proportion was increased to more than 60%. However, despite the abundance of viral miRNAs during the early phase of infection, the expression of some MCMV miRNAs appeared to be regulated. Hence, for three miRNAs we observed polyuridylation of their 3' end, coupled to subsequent degradation. Individual knockout mutants of two of the most abundant MCMV miRNAs, miR-m01-4 and miR-M44-1, or a double knockout mutant of miR-m21-1 and miR-M23-2, incurred no or only a very mild growth deficit in murine embryonic fibroblasts in vitro.
Collapse
|
152
|
Sheridan BS, Knickelbein JE, Hendricks RL. CD8 T cells and latent herpes simplex virus type 1: keeping the peace in sensory ganglia. Expert Opin Biol Ther 2007; 7:1323-31. [PMID: 17727323 DOI: 10.1517/14712598.7.9.1323] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Herpes simplex virus type 1 (HSV-1) infections represent a significant worldwide heath problem. The lack of an effective therapy to curtail reactivation of HSV-1 from a state of neuronal latency has lead to significant morbidity and mortality. Effective therapies to prevent reactivation must likely elicit a protective CD8 T-cell response that could act to prevent reactivation from sensory neurons prior to release of infectious virus at the periphery. This review focuses on the present understanding of how CD8 T cells maintain HSV-1 latency and how this knowledge could facilitate the generation of more effective therapeutic modalities.
Collapse
Affiliation(s)
- Brian S Sheridan
- University of Pittsburgh, School of Medicine, Graduate Program in Immunology, 203 Lothrop Street, Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
153
|
Ahmed FE. Role of miRNA in carcinogenesis and biomarker selection: a methodological view. Expert Rev Mol Diagn 2007; 7:569-603. [PMID: 17892365 DOI: 10.1586/14737159.7.5.569] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
miRNAs, their involvement in cancer development and their potential to be robust biomarkers of diagnosis, staging, prognosis and response to therapy are reviewed. In small RNA animal biogenesis, miRNA genes in the nucleus are transcribed to generate long primary transcripts (pri-miRNAs), which are first cropped by RNase-III-type enzyme Drosha to release hairpin intermediates (pre-miRNAs) in the nucleus. Pre-miRNA is then exported to the cytoplasm by exportin-5. Following arrival in the cytoplasm, pre-miRNAs are subjected to the second processing step (dicing) to release the mature miRNA duplex, which is then separated: one strand becomes the mature miRNA and the other is degraded. These tiny miRNAs induce messenger degradation, translational repression or both. However, there is no evidence to demonstrate that these two mechanisms exist in the regulation of the same gene. Since a miRNA can target numerous mRNAs, often in combination with other miRNAs, these miRNAs operate a highly complex regulatory network. The specific function in most mammalian miRNAs is unknown. However, data suggest that miRNA genes, approximately 1% of all human genes, regulate protein production for 20-30% or more of all genes. miRNA expression profiles are effective for classifying solid and hematologic human cancers, and have shown great promise for early cancer detection. This is of great importance for effective treatment before the cells metastasize; therefore, tumors can be surgically resected. Computer-based prediction approaches of miRNAs and their targets, and biological validation techniques for ascertaining these predictions, currently play a central role in the discovery of miRNAs and in elucidating their function. Guidelines have been established for the identification and annotation of new miRNAs to distinguish them from other RNAs, especially siRNAs. These guidelines take into account factors such as transcript structure, conservation and processing, and a centralized, searchable database of all possible miRNA sequence information and annotation for humans and of more than 38 other species. Two approaches are used to characterize miRNAs: studying expression of known miRNAs by hybridization-based techniques (e.g., northern blots, RNase protection, primer extension, real-time, quantitative PCR and microarrays) or discovery of novel miRNAs molecules by cloning and sequencing. Owing to their adaptability and high throughput, microarrays may prove to be the preferred platform for whole-genome miRNA expression analysis.
Collapse
Affiliation(s)
- Farid E Ahmed
- East Carolina University, Department of Radiation Oncology, LSB 014, Leo W Jenkins Cancer Center, The Brody School of Medicine, 600 Moye Blvd, LSB 003, Greenville, NC 27858, USA.
| |
Collapse
|
154
|
Huang J, Wang F, Argyris E, Chen K, Liang Z, Tian H, Huang W, Squires K, Verlinghieri G, Zhang H. Cellular microRNAs contribute to HIV-1 latency in resting primary CD4+ T lymphocytes. Nat Med 2007; 13:1241-1247. [PMID: 17906637 DOI: 10.1038/nm1639] [Citation(s) in RCA: 619] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2007] [Accepted: 08/09/2007] [Indexed: 12/11/2022]
Abstract
The latency of human immunodeficiency virus type 1 (HIV-1) in resting primary CD4+ T cells is the major barrier for the eradication of the virus in patients on suppressive highly active antiretroviral therapy (HAART). Even with optimal HAART treatment, replication-competent HIV-1 still exists in resting primary CD4+ T cells. Multiple restriction factors that act upon various steps of the viral life cycle could contribute to viral latency. Here we show that cellular microRNAs (miRNAs) potently inhibit HIV-1 production in resting primary CD4+ T cells. We have found that the 3' ends of HIV-1 messenger RNAs are targeted by a cluster of cellular miRNAs including miR-28, miR-125b, miR-150, miR-223 and miR-382, which are enriched in resting CD4+ T cells as compared to activated CD4+ T cells. Specific inhibitors of these miRNAs substantially counteracted their effects on the target mRNAs, measured either as HIV-1 protein translation in resting CD4+ T cells transfected with HIV-1 infectious clones, or as HIV-1 virus production from resting CD4+ T cells isolated from HIV-1-infected individuals on suppressive HAART. Our data indicate that cellular miRNAs are pivotal in HIV-1 latency and suggest that manipulation of cellular miRNAs could be a novel approach for purging the HIV-1 reservoir.
Collapse
Affiliation(s)
- Jialing Huang
- Center for Human Virology, Division of Infectious Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Skalsky RL, Samols MA, Plaisance KB, Boss IW, Riva A, Lopez MC, Baker HV, Renne R. Kaposi's sarcoma-associated herpesvirus encodes an ortholog of miR-155. J Virol 2007; 81:12836-45. [PMID: 17881434 PMCID: PMC2169101 DOI: 10.1128/jvi.01804-07] [Citation(s) in RCA: 351] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that posttranscriptionally regulate gene expression by binding to 3'-untranslated regions (3'UTRs) of target mRNAs. Kaposi's sarcoma-associated herpesvirus (KSHV), a virus linked to malignancies including primary effusion lymphoma (PEL), encodes 12 miRNA genes, but only a few regulatory targets are known. We found that KSHV-miR-K12-11 shares 100% seed sequence homology with hsa-miR-155, an miRNA frequently found to be up-regulated in lymphomas and critically important for B-cell development. Based on this seed sequence homology, we hypothesized that both miRNAs regulate a common set of target genes and, as a result, could have similar biological activities. Examination of five PEL lines showed that PELs do not express miR-155 but do express high levels of miR-K12-11. Bioinformatic tools predicted the transcriptional repressor BACH-1 to be targeted by both miRNAs, and ectopic expression of either miR-155 or miR-K12-11 inhibited a BACH-1 3'UTR-containing reporter. Furthermore, BACH-1 protein levels are low in cells expressing either miRNA. Gene expression profiling of miRNA-expressing stable cell lines revealed 66 genes that were commonly down-regulated. For select genes, miRNA targeting was confirmed by reporter assays. Thus, based on our in silico predictions, reporter assays, and expression profiling data, miR-K12-11 and miR-155 regulate a common set of cellular targets. Given the role of miR-155 during B-cell maturation, we speculate that miR-K12-11 may contribute to the distinct developmental phenotype of PEL cells, which are blocked in a late stage of B-cell development. Together, these findings indicate that KSHV miR-K12-11 is an ortholog of miR-155.
Collapse
Affiliation(s)
- Rebecca L Skalsky
- Department of Molecular Genetics and Microbiology, and Shands Cancer Center, 1376 Mowry Road, Gainesville, FL 32610-3633, USA
| | | | | | | | | | | | | | | |
Collapse
|
156
|
Abstract
Plants and invertebrates can protect themselves from viral infection through RNA silencing. This antiviral immunity involves production of virus-derived small interfering RNAs (viRNAs) and results in specific silencing of viruses by viRNA-guided effector complexes. The proteins required for viRNA production as well as several key downstream components of the antiviral immunity pathway have been identified in plants, flies, and worms. Meanwhile, viral mechanisms to suppress this small RNA-directed immunity by viruses are being elucidated, thereby illuminating an ongoing molecular arms race that likely impacts the evolution of both viral and host genomes.
Collapse
Affiliation(s)
- Shou-Wei Ding
- Department of Plant Pathology and Microbiology, University of California, Riverside, CA 92521, USA
- Correspondence: (S.W.D.), (O.V.)
| | - Olivier Voinnet
- Institut de Biologie Moléculaire des plantes, CNRS, Université Louis Pasteur, 67084 Strasbourg Cedex, France
- Correspondence: (S.W.D.), (O.V.)
| |
Collapse
|
157
|
Jeyaseelan K, Herath WB, Armugam A. MicroRNAs as therapeutic targets in human diseases. Expert Opin Ther Targets 2007; 11:1119-29. [PMID: 17665982 DOI: 10.1517/14728222.11.8.1119] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
MicroRNAs (miRNAs) are important endogenous regulators of gene expression. The specific regulation at both the transcription and the translation level (inhibition or mRNA degradation) opens an avenue to use these small RNA molecules as potential targets for the development of novel drugs as well as for the diagnosis of several human diseases. Important information about the role of a miRNA in disease can be deduced by mimicking or inhibiting its activity and examining its impact on the phenotype/behaviour of the cell or organism. Modulating the activity of a miRNA is expected to lead to improvement in disease symptoms and this implies that the target miRNA plays an important role in the disease. It is also now possible to develop miRNA-based therapeutic products that can either increase or decrease the levels of proteins in pathophysiological conditions such as cancer, cardiovascular diseases, viral diseases, metabolic disorders and programmed cell death. The commercial potential of miRNA and related drugs is expected to exponentially increase within the next few years, yet there are several areas in miRNA biology and delivery that need to be extensively investigated.
Collapse
Affiliation(s)
- Kandiah Jeyaseelan
- National University of Singapore, Department of Biochemistry, Yong Loo Lin School of Medicine, Singapore.
| | | | | |
Collapse
|
158
|
Carpenter D, Hsiang C, Brown DJ, Jin L, Osorio N, BenMohamed L, Jones C, Wechsler SL. Stable cell lines expressing high levels of the herpes simplex virus type 1 LAT are refractory to caspase 3 activation and DNA laddering following cold shock induced apoptosis. Virology 2007; 369:12-8. [PMID: 17727910 PMCID: PMC2276668 DOI: 10.1016/j.virol.2007.07.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Revised: 06/22/2007] [Accepted: 07/18/2007] [Indexed: 12/19/2022]
Abstract
The herpes simplex virus type 1 (HSV-1) latency associated transcript (LAT) gene's anti-apoptosis activity plays a central, but not fully elucidated, role in enhancing the virus's reactivation phenotype. In transient transfection experiments, LAT increases cell survival following an apoptotic insult in the absence of other HSV-1 genes. However, the high background of untransfected cells has made it difficult to demonstrate that LAT inhibits specific apoptotic factors such as caspases. Here we report that, in mouse neuroblastoma cell lines (C1300) stably expressing high levels of LAT, cold shock induced apoptosis was blocked as judged by increased survival, protection against DNA fragmentation (by DNA ladder assay), and inhibition of caspase 3 cleavage and activation (Western blots). To our knowledge, this is the first report providing direct evidence that LAT blocks two biochemical hallmarks of apoptosis, caspase 3 cleavage and DNA laddering, in the absence of other HSV-1 gene products.
Collapse
Affiliation(s)
- Dale Carpenter
- The Eye Institute, University of California Irvine, School of Medicine, Irvine, CA 92697
| | - Chinhui Hsiang
- The Eye Institute, University of California Irvine, School of Medicine, Irvine, CA 92697
| | - Donald J. Brown
- The Eye Institute, University of California Irvine, School of Medicine, Irvine, CA 92697
| | - Ling Jin
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331
| | - Nelson Osorio
- The Eye Institute, University of California Irvine, School of Medicine, Irvine, CA 92697
| | - Lbachir BenMohamed
- The Cellular and Molecular Immunology Laboratory, The Eye Institute, University of California Irvine, School of Medicine, Irvine, CA 92697
- Center for Immunology, University of California Irvine, Irvine, CA 92697
| | - Clinton Jones
- Department of Veterinary and Biomedical Sciences, University of Nebraska, Lincoln, NE 68583-0905, USA
| | - Steven L. Wechsler
- The Eye Institute, University of California Irvine, School of Medicine, Irvine, CA 92697
- Department of Microbiology and Molecular Genetics, University of California Irvine, School of Medicine, Irvine, CA 92697
- The Center for Virus Research, University of California, Irvine, Irvine, CA 92697
- *Corresponding author: Dr. Steven L. Wechsler., Telephone: 714-456-7362, Fax: 714-456-5073, Mailing address: Steven Wechsler; University of California Irvine Medical Center; Dept of Ophthalmology; 101 The City Drive; Building 55, Room 226; Orange, CA 92868
| |
Collapse
|
159
|
Abstract
In plants and invertebrates, Dicer genes play a critical role against infections by RNA viruses. In this issue, Otsuka et al. (2007) report that Dicer mutant mice are hypersusceptible to infection by the RNA virus VSV.
Collapse
Affiliation(s)
- Stefanie Müller
- Faculté des Sciences de la Vie, Université Louis Pasteur, F-67070 Strasbourg, France
| | | |
Collapse
|
160
|
Burnside J, Morgan RW. Genomics and Marek's disease virus. Cytogenet Genome Res 2007; 117:376-87. [PMID: 17675881 DOI: 10.1159/000103201] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Accepted: 08/12/2006] [Indexed: 12/11/2022] Open
Abstract
Marek's disease virus (MDV), a lymphotrophic alphaherpesvirus of chickens, causes a disease that is characterized by tumor formation, immunosuppression and neurological disorders. Recent developments in chicken genomics have been applied to studies of MDV and have advanced our understanding of both the virus and the disease it causes. We have constructed and used microarrays to identify host genes that are up-regulated in chicken embryo fibroblasts infected with MDV as a first step to catalog the host response to infection. An additional level of gene regulation lies at the level of microRNAs (miRNAs). miRNAs are a class of small (approximately 22 nt) regulatory molecules encoded by a wide variety of organisms, including some viruses, that block translation or induce degradation of specific mRNAs. Herpesviruses, which replicate in the nuclei of infected cells, are a particularly important class of viruses that express miRNAs. miRNAs from two of the oncogenic herpesviruses; namely, Kaposi's sarcoma herpesvirus (KSHV) and Epstein-Barr virus (EBV) have been cataloged. We recently identified MDV-encoded miRNAs. One cluster of miRNAs flanks the meq oncogene, and a second cluster maps to the latency associated transcript (LAT) region of the genome. The LATs are encoded anti-sense to the ICP4 immediate early gene, and the meq gene, which is unique to pathogenic serotypes of MDV, is the most likely oncoprotein or co-oncoprotein encoded by MDV. The conservation of these sequences is suggestive of an important role in pathogenesis.
Collapse
Affiliation(s)
- J Burnside
- Department of Animal and Food Sciences, Delaware Biotechnology Institute, University of Delaware, Newark, DE 19711, USA.
| | | |
Collapse
|
161
|
Carpenter D, Henderson G, Hsiang C, Osorio N, BenMohamed L, Jones C, Wechsler SL. Introducing point mutations into the ATGs of the putative open reading frames of the HSV-1 gene encoding the latency associated transcript (LAT) reduces its anti-apoptosis activity. Microb Pathog 2007; 44:98-102. [PMID: 17900852 PMCID: PMC2291025 DOI: 10.1016/j.micpath.2007.07.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2007] [Indexed: 01/01/2023]
Abstract
The herpes simplex virus type 1 (HSV-1) latency associated transcript (LAT) gene has anti-apoptosis activity that directly or indirectly enhances the virus's reactivation phenotype in small animal models. The first 1.5 kb of the primary 8.3 kb LAT is sufficient and some or all of it is necessary for LAT's anti-apoptosis in transient transfection assays and for LAT's ability to enhance the reactivation phenotype. Based on LAT's genomic sequence, the first 1.5 kb contains eight potential open reading frames (ORFs) defined as an ATG followed by an in frame termination codon. In this study, point mutations were introduced into the ATGs of ORFs present in the 1.5 kb fragment of LAT. Mutagenesis of all eight ATGs in LAT ORFs consistently reduced the anti-apoptotic activity of LAT in transiently transfected mouse neuroblastoma cells regardless of whether apoptosis was induced by caspase 8 or caspase 9. Mutation of the six ATGs located in the stable intron sequences within the 1.5 kb LAT had a dramatic effect on caspase 9, but not caspase 8, induced apoptosis. For both caspase 8 and caspase 9 induced apoptosis, mutating the two ATGs in the exon of the LAT 1.5 kb fragment reduced, but did not eliminate the anti-apoptotic activity of LAT. These studies suggest that altering the fine structure of regulatory RNA or expression of a putative LAT ORF regulates the anti-apoptosis activity of LAT. These studies also indicate that more than one function is present in the 1.5 kb LAT fragment.
Collapse
Affiliation(s)
- Dale Carpenter
- The Eye Institute, University of California Irvine, School of Medicine, Irvine, CA 92697
| | - Gail Henderson
- Department of Veterinary and Biomedical Sciences, Nebraska Center for Virology, University of Nebraska, Lincoln, NE 68583-0905, USA.
| | - Chinhui Hsiang
- The Eye Institute, University of California Irvine, School of Medicine, Irvine, CA 92697
| | - Nelson Osorio
- The Eye Institute, University of California Irvine, School of Medicine, Irvine, CA 92697
| | - Lbachir BenMohamed
- The Cellular and Molecular Immunology Laboratory, The Eye Institute, University of California Irvine, School of Medicine, Irvine, CA 92697.
- Center for Immunology, University of California Irvine, Irvine, CA 92697
| | - Clinton Jones
- Department of Veterinary and Biomedical Sciences, Nebraska Center for Virology, University of Nebraska, Lincoln, NE 68583-0905, USA.
| | - Steven L. Wechsler
- The Eye Institute, University of California Irvine, School of Medicine, Irvine, CA 92697
- Department of Microbiology and Molecular Genetics, University of California Irvine, School of Medicine, Irvine, CA 92697
- The Center for Virus Research, University of California, Irvine, Irvine, CA 92697
- *Corresponding author: Dr. Steven L. Wechsler. Telephone: 714-456-7362, Fax: 714-456-5073,
| |
Collapse
|
162
|
Zhang G, Raghavan B, Kotur M, Cheatham J, Sedmak D, Cook C, Waldman J, Trgovcich J. Antisense transcription in the human cytomegalovirus transcriptome. J Virol 2007; 81:11267-81. [PMID: 17686857 PMCID: PMC2045512 DOI: 10.1128/jvi.00007-07] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Human cytomegalovirus (HCMV) infections are prevalent in human populations and can cause serious diseases, especially in those with compromised or immature immune systems. The HCMV genome of 230 kb is among the largest of the herpesvirus genomes. Although the entire sequence of the laboratory-adapted AD169 strain of HCMV has been available for 18 years, the precise number of viral genes is still in question. We undertook an analysis of the HCMV transcriptome as an approach to enumerate and analyze the gene products of HCMV. Transcripts of HCMV-infected fibroblasts were isolated at different times after infection and used to generate cDNA libraries representing different temporal classes of viral genes. cDNA clones harboring viral sequences were selected and subjected to sequence analysis. Of the 604 clones analyzed, 45% were derived from genomic regions predicted to be noncoding. Additionally, at least 55% of the cDNA clones in this study were completely or partially antisense to known or predicted HCMV genes. The remarkable accumulation of antisense transcripts during infection suggests that currently available genomic maps based on open-reading-frame and other in silico analyses may drastically underestimate the true complexity of viral gene products. These findings also raise the possibility that aspects of both the HCMV life cycle and genome organization are influenced by antisense transcription. Correspondingly, virus-derived noncoding and antisense transcripts may shed light on HCMV pathogenesis and may represent a new class of targets for antiviral therapies.
Collapse
Affiliation(s)
- Guojuan Zhang
- The Ohio State University, Department of Pathology, 4162 Graves Hall, 333 West 10th Avenue, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
163
|
Margolis TP, Elfman FL, Leib D, Pakpour N, Apakupakul K, Imai Y, Voytek C. Spontaneous reactivation of herpes simplex virus type 1 in latently infected murine sensory ganglia. J Virol 2007; 81:11069-74. [PMID: 17686862 PMCID: PMC2045564 DOI: 10.1128/jvi.00243-07] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Careful studies of mouse trigeminal ganglia (TG) latently infected with herpes simplex virus type 1 (HSV-1) indicate the presence of productive cycle viral gene products and persistent immune response, suggesting ongoing spontaneous viral reactivation in these tissues. In the present study we set out to determine whether infectious virus is present in murine TG latently infected with HSV-1 (KOS). At 37 days after ocular inoculation we found a small amount of infectious virus in ca. 6% of latently infected murine TG. Furthermore, the amount of infectious virus that we detected (PFU per viral antigen-positive neuron) was similar to that detected in acutely infected ganglia. We conclude that spontaneous reactivation of infectious HSV-1 occurs in the mouse TG and is likely the principle cause of viral protein expression in these tissues. We next examined the role of latency-associated transcript (LAT) in spontaneous ganglionic reactivation by examining ganglia latently infected with KOS dlLAT1.8, a LAT deletion virus. Through the use of immunocytochemistry we found that KOS dlLAT1.8 had a rate of spontaneous ganglionic reactivation very similar to that of HSV-1 (KOS). Studying spontaneous ganglionic reactivation of HSV in the mouse TG allows a direct study of viral reactivation from latently infected neurons without the potential confounders and complicating downstream events that accompany the study of viral reactivation by explantation or peripheral viral shedding. Since most cases of human viral shedding and reactivation are not associated with a known trigger, spontaneous ganglionic reactivation of HSV-1 may be a better model of human disease than existing models.
Collapse
Affiliation(s)
- Todd P Margolis
- Francis I. Proctor Foundation, UCSF, Medical Sciences Building S-310, 513 Parnassus Ave., San Francisco, CA 94143-0412, USA.
| | | | | | | | | | | | | |
Collapse
|
164
|
Affiliation(s)
- Bryan R Cullen
- Center for Virology and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27707, USA.
| |
Collapse
|
165
|
Goodrum F, Reeves M, Sinclair J, High K, Shenk T. Human cytomegalovirus sequences expressed in latently infected individuals promote a latent infection in vitro. Blood 2007; 110:937-45. [PMID: 17440050 PMCID: PMC1924770 DOI: 10.1182/blood-2007-01-070078] [Citation(s) in RCA: 193] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Accepted: 04/05/2007] [Indexed: 01/20/2023] Open
Abstract
Latency enables human cytomegalovirus (HCMV) to persist in the hematopoietic cells of infected individuals indefinitely and prevents clearance of the pathogen. Despite its critical importance to the viral infectious cycle, viral mechanisms that contribute to latency have not been identified. We compared the ability of low-passage clinical and laboratory-adapted strains of HCMV to establish a latent infection in primary human CD34(+) cells. The low-passage strains, Toledo and FIX, established an infection with the hallmarks of latency, whereas the laboratory strains, AD169 and Towne, replicated producing progeny virus. We hypothesized that ULb' region of the genome, which is unique to low-passage strains, may encode a latency-promoting activity. We created and analyzed recombinant viruses lacking segments or individual open reading frames (ORFs) in the ULb' region. One 5-kb segment, and more specifically the UL138 ORF, was required for HCMV to establish and/or maintain a latent infection in hematopoietic progenitor cells infected in vitro. This is the first functional demonstration of a virus-coded sequence required for HCMV latency. Importantly, UL138 RNA was expressed in CD34(+) cells and monocytes from HCMV-seropositive, healthy individuals. UL138 might be a target for antivirals against latent virus.
Collapse
Affiliation(s)
- Felicia Goodrum
- Department of Immunobiology, BIO5 Institute, University of Arizona, Tucson, AZ 85711, USA.
| | | | | | | | | |
Collapse
|
166
|
Randall G, Panis M, Cooper JD, Tellinghuisen TL, Sukhodolets KE, Pfeffer S, Landthaler M, Landgraf P, Kan S, Lindenbach BD, Chien M, Weir DB, Russo JJ, Ju J, Brownstein MJ, Sheridan R, Sander C, Zavolan M, Tuschl T, Rice CM. Cellular cofactors affecting hepatitis C virus infection and replication. Proc Natl Acad Sci U S A 2007; 104:12884-9. [PMID: 17616579 PMCID: PMC1937561 DOI: 10.1073/pnas.0704894104] [Citation(s) in RCA: 435] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Indexed: 02/06/2023] Open
Abstract
Recently identified hepatitis C virus (HCV) isolates that are infectious in cell culture provide a genetic system to evaluate the significance of virus-host interactions for HCV replication. We have completed a systematic RNAi screen wherein siRNAs were designed that target 62 host genes encoding proteins that physically interact with HCV RNA or proteins or belong to cellular pathways thought to modulate HCV infection. This includes 10 host proteins that we identify in this study to bind HCV NS5A. siRNAs that target 26 of these host genes alter infectious HCV production >3-fold. Included in this set of 26 were siRNAs that target Dicer, a principal component of the RNAi silencing pathway. Contrary to the hypothesis that RNAi is an antiviral pathway in mammals, as has been reported for subgenomic HCV replicons, siRNAs that target Dicer inhibited HCV replication. Furthermore, siRNAs that target several other components of the RNAi pathway also inhibit HCV replication. MicroRNA profiling of human liver, human hepatoma Huh-7.5 cells, and Huh-7.5 cells that harbor replicating HCV demonstrated that miR-122 is the predominant microRNA in each environment. miR-122 has been previously implicated in positively regulating the replication of HCV genotype 1 replicons. We find that 2'-O-methyl antisense oligonucleotide depletion of miR-122 also inhibits HCV genotype 2a replication and infectious virus production. Our data define 26 host genes that modulate HCV infection and indicate that the requirement for functional RNAi for HCV replication is dominant over any antiviral activity this pathway may exert against HCV.
Collapse
Affiliation(s)
- Glenn Randall
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, and
- Department of Microbiology, University of Chicago, Chicago, IL 60637
| | - Maryline Panis
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, and
| | - Jacob D. Cooper
- Department of Microbiology, University of Chicago, Chicago, IL 60637
| | | | - Karen E. Sukhodolets
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Sebastien Pfeffer
- Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY 10021
| | - Markus Landthaler
- Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY 10021
| | - Pablo Landgraf
- Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY 10021
| | - Sherry Kan
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, and
| | - Brett D. Lindenbach
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, and
| | | | - David B. Weir
- Department of Chemical Engineering, Columbia University, New York, NY 10027
| | | | - Jingyue Ju
- Columbia Genome Center, New York, NY 10032
- Department of Chemical Engineering, Columbia University, New York, NY 10027
| | | | - Robert Sheridan
- Computational Biology Center, Memorial Sloan–Kettering Cancer Center, New York, NY 10021; and
| | - Chris Sander
- Computational Biology Center, Memorial Sloan–Kettering Cancer Center, New York, NY 10021; and
| | - Mihaela Zavolan
- Biozentrum, Universität Basel, CH-4056 Basel, Switzerland; and
| | - Thomas Tuschl
- Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY 10021
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, and
| |
Collapse
|
167
|
Xu N, Segerman B, Zhou X, Akusjärvi G. Adenovirus virus-associated RNAII-derived small RNAs are efficiently incorporated into the rna-induced silencing complex and associate with polyribosomes. J Virol 2007; 81:10540-9. [PMID: 17652395 PMCID: PMC2045446 DOI: 10.1128/jvi.00885-07] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Adenovirus type 5 encodes two highly structured short RNAs, the virus-associated (VA) RNAI and RNAII. Both are processed by Dicer into small RNAs that are incorporated into the RNA-induced silencing complex (RISC). We show here, by cloning of small RNAs, that approximately 80% of Ago2-containing RISC immunopurified from late-infected cells is associated with VA RNA-derived small RNAs (mivaRNAs). Most surprisingly, VA RNAII, which is expressed at 20-fold lower levels compared to that of VA RNAI, appears to be the preferred substrate for Dicer and accounts for approximately 60% of all small RNAs in RISC. The mivaRNAs are derived from the 3' strand of the terminal stems of the VA RNAs, with the major fraction of VA RNAII starting at position 138. The small RNAs derived from VA RNAI were more heterogeneous in size, with the two predominant small RNAs starting at positions 137 and 138. Collectively, our results suggest that the mivaRNAs are efficiently used for RISC assembly in late-infected cells. Potentially, they function as miRNAs, regulating translation of cellular mRNAs. In support of this hypothesis, we detected a fraction of the VA RNAII-derived mivaRNAs on polyribosomes.
Collapse
Affiliation(s)
- Ning Xu
- Department of Medical Biochemistry and Microbiology, Uppsala Biomedical Center, Husargatan 3, S-751 23 Uppsala, Sweden
| | | | | | | |
Collapse
|
168
|
Nguyen ML, Kraft RM, Blaho JA. Susceptibility of cancer cells to herpes simplex virus-dependent apoptosis. J Gen Virol 2007; 88:1866-1875. [PMID: 17554017 DOI: 10.1099/vir.0.82868-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Apoptosis has recently been associated with herpes simplex virus 1 (HSV-1) latency and disease severity. There is an intricate balance between pro- and anti-apoptotic processes during HSV-1 infection. When anti-apoptotic pathways are suppressed, this balance is upset and the cells die by apoptosis, referred to here as HSV-1-dependent apoptosis (HDAP). It has been observed previously that HeLa cancer cells exhibit an enhanced sensitivity to HDAP. Here, a series of specific patient-derived cancer cells was utilized to investigate the cell-type specificity of HDAP. The results showed that a human mammary tumour cell line was sensitive to HDAP, whilst syngeneic normal cells were resistant. Furthermore, low-passage-number primary human mammary epithelial cells were resistant to HDAP. When the susceptibility of human colon, brain, breast and cervical cancer cells was assessed, the only cells insensitive to HDAP were those resistant to all environmental stimuli tested. This implies that the HDAP resistance was probably due to mutations in the cellular apoptotic machinery. Thus, the susceptibility of cancer cells to HDAP requires that they possess a functional ability to undergo programmed cell death.
Collapse
Affiliation(s)
- Marie L Nguyen
- Department of Microbiology, One Gustave L. Levy Place, Mount Sinai School of Medicine, New York, NY 10029-6574, USA
| | - Rachel M Kraft
- Department of Microbiology, One Gustave L. Levy Place, Mount Sinai School of Medicine, New York, NY 10029-6574, USA
| | - John A Blaho
- Department of Microbiology, One Gustave L. Levy Place, Mount Sinai School of Medicine, New York, NY 10029-6574, USA
| |
Collapse
|
169
|
Stern-Ginossar N, Elefant N, Zimmermann A, Wolf DG, Saleh N, Biton M, Horwitz E, Prokocimer Z, Prichard M, Hahn G, Goldman-Wohl D, Greenfield C, Yagel S, Hengel H, Altuvia Y, Margalit H, Mandelboim1 O. Host immune system gene targeting by a viral miRNA. Science 2007; 317:376-381. [PMID: 17641203 PMCID: PMC4283197 DOI: 10.1126/science.1140956] [Citation(s) in RCA: 510] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Virally encoded microRNAs (miRNAs) have recently been discovered in herpesviruses. However, their biological roles are mostly unknown. We developed an algorithm for the prediction of miRNA targets and applied it to human cytomegalovirus miRNAs, resulting in the identification of the major histocompatibility complex class I-related chain B (MICB) gene as a top candidate target of hcmv-miR-UL112. MICB is a stress-induced ligand of the natural killer (NK) cell activating receptor NKG2D and is critical for the NK cell killing of virus-infected cells and tumor cells. We show that hcmv-miR-UL112 specifically down-regulates MICB expression during viral infection, leading to decreased binding of NKG2D and reduced killing by NK cells. Our results reveal a miRNA-based immunoevasion mechanism that appears to be exploited by human cytomegalovirus.
Collapse
Affiliation(s)
- Noam Stern-Ginossar
- Lautenberg Center for General and Tumor Immunology, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Naama Elefant
- Department of Molecular Genetics and Biotechnology, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Albert Zimmermann
- Institute for Virology, Heinrich Heine University, D40225 Düsseldorf, Germany
| | - Dana G. Wolf
- Department of Clinical Microbiology and Infectious Diseases, Hadassah University Hospital, Jerusalem, Israel
| | - Nivin Saleh
- Department of Clinical Microbiology and Infectious Diseases, Hadassah University Hospital, Jerusalem, Israel
| | - Moshe Biton
- Lautenberg Center for General and Tumor Immunology, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Elad Horwitz
- Lautenberg Center for General and Tumor Immunology, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Zafnat Prokocimer
- Lautenberg Center for General and Tumor Immunology, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Mark Prichard
- Department of Pediatrics, University of Alabama, Birmingham, AL 35233, USA
| | - Gabriele Hahn
- Max von Pettenkofer Institut, Department of Virology, D80336 Munich, Germany
| | - Debra Goldman-Wohl
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Hospital Mount Scopus, Jerusalem, Israel
| | - Caryn Greenfield
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Hospital Mount Scopus, Jerusalem, Israel
| | - Simcha Yagel
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Hospital Mount Scopus, Jerusalem, Israel
| | - Hartmut Hengel
- Institute for Virology, Heinrich Heine University, D40225 Düsseldorf, Germany
| | - Yael Altuvia
- Department of Molecular Genetics and Biotechnology, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Hanah Margalit
- Department of Molecular Genetics and Biotechnology, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Ofer Mandelboim1
- To whom correspondence should be addressed. (Y.A.); (H.M.); (O.M.)
| |
Collapse
|
170
|
Berkhout B, Jeang KT. RISCy business: MicroRNAs, pathogenesis, and viruses. J Biol Chem 2007; 282:26641-26645. [PMID: 17627941 DOI: 10.1074/jbc.r700023200] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Ben Berkhout
- Acdemisch Medisch Centrum, Meibergdreef 15, 1105 AZ Amsterdam, the Netherlands
| | - Kuan-Teh Jeang
- Laboratory of Molecular Microbiology, NIAID, National Institutes of Health, Bethesda, Maryland 20892.
| |
Collapse
|
171
|
Xing L, Kieff E. Epstein-Barr virus BHRF1 micro- and stable RNAs during latency III and after induction of replication. J Virol 2007; 81:9967-75. [PMID: 17626073 PMCID: PMC2045418 DOI: 10.1128/jvi.02244-06] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Epstein-Barr virus (EBV) microRNAs miR-BHRF1-1, -2, and -3 have been detected in latency III-infected lymphoblasts, where they are encoded within EBNA transcripts (X. Cai, A. Schafer, S. Lu, J. P. Bilello, R. C. Desrosiers, R. Edwards, N. Raab-Traub, and B. R. Cullen, PLoS Pathog. 2:e23, 2006). In latency III-infected lymphoblasts, we have also identified a stable 1.3-kb RNA, which begins 3' to miR-BHRF1-1, includes the BHRF1 open reading frame, and ends near miR-BHRF1-2. This 1.3-kb RNA is the residue of Drosha cleavage of the BHRF1 microRNAs from EBNA transcripts. Early after induction of EBV replication in latency I-infected Akata lymphoblasts, BHRF1 spliced 1.4-kb mRNA accumulated along with low levels of miR-BHRF1-2 and -3 and a 0.9-kb Drosha or miR-BHRF1-2 cleavage product of BHRF1 mRNA. The turning on of latency III infection at 48 to 72 h after induction of EBV replication was associated with higher miR-BHRF1-1, -2, and -3 levels; accumulation of the 1.3-kb RNA residue in the nucleus; abundant BHRF1 spliced 1.4-kb mRNA in the cytoplasm; and more abundant 0.9-kb mRNA cleavage product in the cytoplasm. These findings implicate miR-BHRF1-2 in 3' cleavage of BHRF1 mRNA in the cytoplasm and Drosha in cleavage of latency III EBNA and EBV replication-associated BHRF1 transcripts in the nucleus.
Collapse
Affiliation(s)
- Li Xing
- Brigham and Women's Hospital, Harvard Medical School, Channing Laboratory, 181 Longwood Avenue, Boston, MA 02115-5804, USA
| | | |
Collapse
|
172
|
Jin L, Perng GC, Carpenter D, Mott KR, Osorio N, Naito J, Brick DJ, Jones C, Wechsler SL. Reactivation phenotype in rabbits of a herpes simplex virus type 1 mutant containing an unrelated antiapoptosis gene in place of latency-associated transcript. J Neurovirol 2007; 13:78-84. [PMID: 17454452 DOI: 10.1080/13550280601164333] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Latency-associated transcript (LAT) significantly enhances the spontaneous reactivation phenotype of herpes simplex virus type 1 (HSV-1). The mechanism by which LAT accomplishes this has been elusive. To determine if LAT's antiapoptosis activity is involved, the authors used a rabbit eye model to analyze the spontaneous reactivation phenotype of an HSV-1 mutant in which LAT was replaced by an unrelated antiapoptosis gene. This virus, dLAT-cpIAP, contains the open reading frame of the baculovirus inhibitor of apoptosis protein gene (cpIAP) in place of LAT, under control of the LAT promoter. The authors report here that in a rabbit ocular model of infection, dLAT-cpIAP had a spontaneous reactivation phenotype similar to wild-type virus and significantly higher than LAT(-) viruses. This was consistent with their previous findings using the mouse trigeminal ganglia explant-induced reactivation model. Whether LAT (and in the case of dLAT-cpIAP, cpIAP) enhances the spontaneous reactivation phenotype by functioning during establishment of latency, maintenance of latency, or reactivation from latency, or during two or more of these periods, remains to be determined. Regardless, the results presented in this study strongly support the hypothesis that LAT's antiapoptosis activity is the dominant function that enhances HSV-1's spontaneous reactivation phenotype.
Collapse
Affiliation(s)
- Ling Jin
- The Eye Institute, University of California Irvine, School of Medicine, Irvine, California 92697, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Abstract
MicroRNAs (miRNAs) are an important class of small, noncoding, regulatory RNAs found to be involved in regulating a wide variety of important cellular processes by the sequence-specific inhibition of gene expression. Viruses have evolved a number of mechanisms to take advantage of the regulatory potential of this highly conserved, ubiquitous pathway known as RNA interference (RNAi). This review will focus on the recent efforts to understand the complex relationship between vertebrate viruses and the RNAi pathway, as well as the role of silencing pathways in the inhibition of pathogenic genetic elements, including transposons and retrotransposons.
Collapse
Affiliation(s)
- Derek M Dykxhoorn
- The CBR Institute for Biomedical Research, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
174
|
Kari I, Syrjänen S, Johansson B, Peri P, He B, Roizman B, Hukkanen V. Antisense RNA directed to the human papillomavirus type 16 E7 mRNA from herpes simplex virus type 1 derived vectors is expressed in CaSki cells and downregulates E7 mRNA. Virol J 2007; 4:47. [PMID: 17547759 PMCID: PMC1892547 DOI: 10.1186/1743-422x-4-47] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Accepted: 06/04/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human papillomavirus (HPV) infection is known to be the most important etiologic factor of cervical cancer. There is no HPV specific therapy available for treatment of invasive squamous cell carcinoma of the cervix and its precursor lesions. The present study elucidates the potential to use herpes simplex virus (HSV) derived vectors for expression of antisense RNA to HPV -16 E7 oncogene. RESULTS We have constructed replication competent, nonneuroinvasive HSV-1 vectors, deleted of the gamma134.5 gene. The vectors express RNA antisense to the first 100 nucleotides of the HPV-16 E7 gene. We assayed the ability of the antisense E7 vectors R5225 (tk-) and R5226 (tk+), to produce antisense RNA, as well as the consequent effects on E7 mRNA and protein levels in HPV-16 positive CaSki cells. Anti-E7 RNA was expressed by both constructs in a dose-dependent manner. Expression of HPV-16 E7 mRNA was downregulated effectively in CaSki cells infected with the tk- recombinant R5225 or with R5226. The tk+ recombinant R5226 was effective in downregulating E7 protein expression. CONCLUSION We have shown that anti-E7 RNA expressed from an HSV vector could efficiently downregulate HPV-16 E7 mRNA and E7 protein expression in CaSki cells. We conclude that HSV vectors may become a useful tool for gene therapy of HPV infections.
Collapse
MESH Headings
- Antiviral Agents/pharmacology
- Cell Line, Tumor
- Down-Regulation
- Gene Expression/drug effects
- Gene Expression Regulation, Viral
- Genetic Vectors/genetics
- Herpesvirus 1, Human/genetics
- Human papillomavirus 16/genetics
- Humans
- Oncogene Proteins, Viral/biosynthesis
- Oncogene Proteins, Viral/genetics
- Papillomavirus E7 Proteins
- RNA, Antisense/biosynthesis
- RNA, Antisense/genetics
- RNA, Antisense/pharmacology
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/biosynthesis
- RNA, Viral/antagonists & inhibitors
- RNA, Viral/biosynthesis
Collapse
Affiliation(s)
- Ilkka Kari
- Department of Virology, Institute of Dentistry, University of Turku, Turku, Finland
- MediCity Research Laboratory, Institute of Dentistry, University of Turku, Turku, Finland
- Department of Oral Pathology, Institute of Dentistry, University of Turku, Turku, Finland
| | - Stina Syrjänen
- MediCity Research Laboratory, Institute of Dentistry, University of Turku, Turku, Finland
- Department of Oral Pathology, Institute of Dentistry, University of Turku, Turku, Finland
| | - Bo Johansson
- MediCity Research Laboratory, Institute of Dentistry, University of Turku, Turku, Finland
- Department of Oral Pathology, Institute of Dentistry, University of Turku, Turku, Finland
- Department of Clinical Virology, Karolinska University Hospital, Stockholm, Sweden
| | - Piritta Peri
- Department of Virology, Institute of Dentistry, University of Turku, Turku, Finland
| | - Bin He
- The Marjorie B. Kovler Viral Oncology Laboratories, The University of Chicago, Chicago, IL, USA
- Department of Microbiology and Immunology, University of Illinois, Chicago, IL, USA
| | - Bernard Roizman
- The Marjorie B. Kovler Viral Oncology Laboratories, The University of Chicago, Chicago, IL, USA
| | - Veijo Hukkanen
- Department of Virology, Institute of Dentistry, University of Turku, Turku, Finland
- MediCity Research Laboratory, Institute of Dentistry, University of Turku, Turku, Finland
- Department of Microbiology, University of Oulu, Oulu, Finland
| |
Collapse
|
175
|
Kapranov P, Willingham AT, Gingeras TR. Genome-wide transcription and the implications for genomic organization. Nat Rev Genet 2007; 8:413-23. [PMID: 17486121 DOI: 10.1038/nrg2083] [Citation(s) in RCA: 546] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent evidence of genome-wide transcription in several species indicates that the amount of transcription that occurs cannot be entirely accounted for by current sets of genome-wide annotations. Evidence indicates that most of both strands of the human genome might be transcribed, implying extensive overlap of transcriptional units and regulatory elements. These observations suggest that genomic architecture is not colinear, but is instead interleaved and modular, and that the same genomic sequences are multifunctional: that is, used for multiple independently regulated transcripts and as regulatory regions. What are the implications and consequences of such an interleaved genomic architecture in terms of increased information content, transcriptional complexity, evolution and disease states?
Collapse
Affiliation(s)
- Philipp Kapranov
- Affymetrix, Inc., 3420 Central Expressway, Santa Clara, California 95051, USA
| | | | | |
Collapse
|
176
|
Wood VHJ, O'Neil JD, Wei W, Stewart SE, Dawson CW, Young LS. Epstein-Barr virus-encoded EBNA1 regulates cellular gene transcription and modulates the STAT1 and TGFbeta signaling pathways. Oncogene 2007; 26:4135-47. [PMID: 17486072 DOI: 10.1038/sj.onc.1210496] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The Epstein-Barr virus (EBV)-encoded EBNA1 protein is expressed in all virus-associated tumors where it plays an essential role in the maintenance, replication and transcription of the EBV genome. Transcriptional profiling of EBNA1-expressing carcinoma cells demonstrated that EBNA1 also influences the expression of a range of cellular genes including those involved in translation, transcription and cell signaling. Of particular interest was the ability of EBNA1 to enhance expression of STAT1 and sensitize cells to interferon-induced STAT1 activation with resultant enhancement of major histocompatibility complex expression. A negative effect of EBNA1 on the expression of TGFbeta1-responsive betaig-h3 and PAI-1 genes was confirmed at the protein level in EBV-infected carcinoma cells. This effect resulted from the ability of EBNA1 to repress TGFbeta1-induced transcription via a reduction in the interaction of SMAD2 with SMAD4. More detailed analysis revealed that EBNA1 induces a lower steady-state level of SMAD2 protein as a consequence of increased protein turnover. These data show that EBNA1 can influence cellular gene transcription resulting in effects that may contribute to the development of EBV-associated tumors.
Collapse
Affiliation(s)
- V H J Wood
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Edgbaston, Birmingham, UK
| | | | | | | | | | | |
Collapse
|
177
|
Yao Y, Zhao Y, Xu H, Smith LP, Lawrie CH, Sewer A, Zavolan M, Nair V. Marek's disease virus type 2 (MDV-2)-encoded microRNAs show no sequence conservation with those encoded by MDV-1. J Virol 2007; 81:7164-70. [PMID: 17459919 PMCID: PMC1933330 DOI: 10.1128/jvi.00112-07] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
MicroRNAs (miRNAs) are increasingly being recognized as major regulators of gene expression in many organisms, including viruses. Among viruses, members of the family Herpesviridae account for the majority of the currently known virus-encoded miRNAs. The highly oncogenic Marek's disease virus type 1 (MDV-1), an avian herpesvirus, has recently been shown to encode eight miRNAs clustered in the MEQ and LAT regions of the viral genome. The genus Mardivirus, to which MDV-1 belongs, also includes the nononcogenic but antigenically related MDV-2. As MDV-1 and MDV-2 are evolutionarily very close, we sought to determine if MDV-2 also encodes miRNAs. For this, we cloned, sequenced, and analyzed a library of small RNAs from the lymphoblastoid cell line MSB-1, previously shown to be coinfected with both MDV-1 and MDV-2. Among the 5,099 small RNA sequences determined from the library, we identified 17 novel MDV-2-specific miRNAs. Out of these, 16 were clustered in a 4.2-kb long repeat region that encodes R-LORF2 to R-LORF5. The single miRNA outside the cluster was located in the short repeat region, within the C-terminal region of the ICP4 homolog. The expression of these miRNAs in MSB-1 cells and infected chicken embryo fibroblasts was further confirmed by Northern blotting analysis. The identification of miRNA clusters within the repeat regions of MDV-2 demonstrates conservation of the relative genomic positions of miRNA clusters in MDV-1 and MDV-2, despite the lack of sequence homology among the miRNAs of the two viruses. The identification of these novel miRNAs adds to the growing list of virus-encoded miRNAs.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cell Line
- Cells, Cultured
- Chick Embryo
- Cloning, Molecular
- Evolution, Molecular
- Fibroblasts/metabolism
- Fibroblasts/virology
- Genome, Viral
- Herpesvirus 2, Gallid/genetics
- Herpesvirus 2, Gallid/metabolism
- Herpesvirus 3, Gallid/genetics
- Herpesvirus 3, Gallid/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Molecular Sequence Data
- Multigene Family
- RNA, Viral/genetics
- RNA, Viral/metabolism
Collapse
Affiliation(s)
- Yongxiu Yao
- Avian Oncogenic Virus Group, Institute for Animal Health, Compton, Berkshire RG20 7NN, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
178
|
Schäfer A, Cai X, Bilello JP, Desrosiers RC, Cullen BR. Cloning and analysis of microRNAs encoded by the primate gamma-herpesvirus rhesus monkey rhadinovirus. Virology 2007; 364:21-7. [PMID: 17451774 PMCID: PMC1941761 DOI: 10.1016/j.virol.2007.03.029] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 02/21/2007] [Accepted: 03/16/2007] [Indexed: 11/30/2022]
Abstract
Several pathogenic human herpesviruses have recently been shown to express virally encoded microRNAs in infected cells. Although the function of these microRNAs is largely unknown, they are hypothesized to play a role in mediating viral replication by downregulating cellular mRNAs encoding antiviral factors. Here, we report the cloning and analysis of microRNAs encoded by Rhesus Monkey Rhadinovirus (RRV), an animal virus model for the pathogenic human gamma-herpesvirus Kaposi's Sarcoma-Associated Herpesvirus (KSHV). RRV expresses several microRNAs that are encoded in the same genomic location as the previously reported KSHV microRNAs, yet these microRNAs are unrelated in primary sequence. These data set the stage for the mutational ablation and phenotypic analysis of RRV mutants lacking one or more viral microRNAs.
Collapse
Affiliation(s)
- Alexandra Schäfer
- Department of Molecular Genetics and Microbiology and Center for Virology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xuezhong Cai
- Department of Molecular Genetics and Microbiology and Center for Virology, Duke University Medical Center, Durham, NC 27710, USA
| | - John P. Bilello
- New England Primate Research Center, Harvard Medical School, Southborough, MA 01772, USA
| | - Ronald C. Desrosiers
- New England Primate Research Center, Harvard Medical School, Southborough, MA 01772, USA
| | - Bryan R. Cullen
- Department of Molecular Genetics and Microbiology and Center for Virology, Duke University Medical Center, Durham, NC 27710, USA
- * Corresponding author. Fax: 919-681-8979, E-mail address: (B. R. Cullen)
| |
Collapse
|
179
|
Geenen K, Nauwynck HJ, De Regge N, Braeckmans K, Favoreel HW. Brn-3a suppresses pseudorabies virus-induced cell death in sensory neurons. J Gen Virol 2007; 88:743-747. [PMID: 17325346 DOI: 10.1099/vir.0.82674-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sensory neurons of the trigeminal ganglion (TG) are of crucial importance in the pathogenesis of many alphaherpesviruses, constituting major target cells for latency and reactivation events. We showed earlier that a subpopulation of porcine TG neurons, in contrast to other porcine cell types, is highly resistant to cell death induced by infection with the porcine alphaherpesvirus pseudorabies virus (PRV). Here, we report that expression of Brn-3a, a neuron-specific transcription factor implicated in cell survival of sensory neurons, correlates with the increased resistance of TG neurons towards PRV-induced cell death. In addition, overexpression of Brn-3a in the sensory neuronal cell line ND7 markedly increased resistance of these cells to PRV-induced cell death. Hence, Brn-3a may play a hitherto uncharacterized role in protection of sensory neurons from alphaherpesvirus-induced cell death, which may have implications for different aspects of the alphaherpesvirus life cycle, including latency/reactivation events.
Collapse
Affiliation(s)
- Kristin Geenen
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Hans J Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Nick De Regge
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Herman W Favoreel
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|
180
|
Gammell P. MicroRNAs: recently discovered key regulators of proliferation and apoptosis in animal cells : Identification of miRNAs regulating growth and survival. Cytotechnology 2007; 53:55-63. [PMID: 19003190 PMCID: PMC2267611 DOI: 10.1007/s10616-007-9049-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Accepted: 01/25/2007] [Indexed: 12/19/2022] Open
Abstract
The relatively recent discovery of miRNAs has added a completely new dimension to the study of the regulation of gene expression. The mechanism of action of miRNAs, the conservation between diverse species and the fact that each miRNA can regulate a number of targets and phenotypes clearly indicates the importance of these molecules. In this review the current state of knowledge relating to miRNA expression and gene regulation is presented, outlining the key morphological and biochemical features controlled by miRNAs with particular emphasis on the key phenotypes that impact on cell growth in bioreactors, namely proliferation and apoptosis.
Collapse
Affiliation(s)
- Patrick Gammell
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin, 11, Ireland,
| |
Collapse
|
181
|
Esau CC, Monia BP. Therapeutic potential for microRNAs. Adv Drug Deliv Rev 2007; 59:101-14. [PMID: 17462786 DOI: 10.1016/j.addr.2007.03.007] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2006] [Accepted: 03/04/2007] [Indexed: 12/19/2022]
Abstract
MiRNAs are a conserved class of non-coding RNAs that negatively regulate gene expression post-transcriptionally. Although their biological roles are largely unknown, examples of their importance in cancer, metabolic disease, and viral infection are accumulating, suggesting that they represent a new class of drug targets in these and likely many other therapeutic areas. Antisense oligonucleotide approaches for inhibiting miRNA function and siRNA-like technologies for replacement of miRNAs are currently being explored as tools for uncovering miRNA biology and as potential therapeutic agents. The next few years should see significant progress in our understanding of miRNA biology and the advancement of the technology for therapeutic modulation of miRNA activity.
Collapse
Affiliation(s)
- Christine C Esau
- Isis Pharmaceuticals, 1896 Rutherford Road, Carlsbad, California 92008, USA.
| | | |
Collapse
|
182
|
Gilden DH, Mahalingam R, Cohrs RJ, Tyler KL. Herpesvirus infections of the nervous system. ACTA ACUST UNITED AC 2007; 3:82-94. [PMID: 17279082 DOI: 10.1038/ncpneuro0401] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Accepted: 11/30/2006] [Indexed: 11/08/2022]
Abstract
There are eight human herpesviruses (HHVs). Primary infection by any of the eight viruses, usually occurring in childhood, is either asymptomatic or produces fever and rash of skin or mucous membranes; other organs might be involved on rare occasions. After primary infection, the virus becomes latent in ganglia or lymphoid tissue. With the exception of HHV-8, which causes Kaposi's sarcoma in patients with AIDS, reactivation of HHVs can produce one or more of the following complications: meningitis, encephalitis, myelitis, vasculopathy, ganglioneuritis, retinal necrosis and optic neuritis. Disease can be monophasic, recurrent or chronic. Infection with each herpesvirus produces distinctive clinical features and imaging abnormalities. This Review highlights the patterns of neurological symptoms and signs, along with the typical imaging abnormalities, produced by each of the HHVs. Optimal virological studies of blood, cerebrospinal fluid and affected tissue for confirmation of diagnosis are discussed; this is particularly important because some HHV infections of the nervous system can be treated successfully with antiviral agents.
Collapse
Affiliation(s)
- Donald H Gilden
- Department of Neurology, University of Colorado Health Sciences Center, Denver, CO 80262, USA.
| | | | | | | |
Collapse
|
183
|
Verjans GMGM, Hintzen RQ, van Dun JM, Poot A, Milikan JC, Laman JD, Langerak AW, Kinchington PR, Osterhaus ADME. Selective retention of herpes simplex virus-specific T cells in latently infected human trigeminal ganglia. Proc Natl Acad Sci U S A 2007; 104:3496-501. [PMID: 17360672 PMCID: PMC1805572 DOI: 10.1073/pnas.0610847104] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Indexed: 11/18/2022] Open
Abstract
Primary infection with herpes simplex virus 1 (HSV-1) and varicella zoster virus (VZV) results in lifelong latent infections of neurons in sensory ganglia such as the trigeminal ganglia (TG). It has been postulated that T cells retained in TG inhibit reactivation of latent virus. The acquisition of TG specimens of individuals within hours after death offered the unique opportunity to characterize the phenotype and specificity of TG-resident T cells in humans. High numbers of activated CD8(+) T cells expressing a late effector memory phenotype were found to reside in latently infected TG. The T cell infiltrate was oligoclonal, and T cells selectively clustered around HSV-1 but not VZV latently infected neurons. Neuronal damage was not observed despite granzyme B expression by the neuron-interacting CD8(+) T cells. The TG-resident T cells, mainly CD8(+) T cells, were directed against HSV-1 and not to VZV, despite neuronal expression of VZV proteins. The results implicate that herpesvirus latency in human TG is associated with a local, persistent T cell response, comprising activated late effector memory CD8(+) T cells that appear to control HSV-1 latency by noncytolytic pathways. In contrast, T cells do not seem to be directly involved in controlling VZV latency in human TG.
Collapse
Affiliation(s)
- Georges M G M Verjans
- Department of Virology, Erasmus Medical Center, Dr. Molewaterplein 50, 3015 GE, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Adams BD, Furneaux H, White BA. The micro-ribonucleic acid (miRNA) miR-206 targets the human estrogen receptor-alpha (ERalpha) and represses ERalpha messenger RNA and protein expression in breast cancer cell lines. Mol Endocrinol 2007; 21:1132-47. [PMID: 17312270 DOI: 10.1210/me.2007-0022] [Citation(s) in RCA: 357] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Micro-RNAs are small noncoding RNAs, which diminish the stability and/or translation of mRNAs. This study examined whether miR-206, previously shown to be elevated in estrogen receptor (ER)alpha-negative breast cancer, regulates the expression of ERalpha. Two putative miR-206 sites, (hERalpha1 and hERalpha2), were found in silico within the 3'-untranslated region of human ERalpha mRNA. Transfection of MCF-7 cells with pre-miR-206 or 2'-O-methyl antagomiR-206 specifically decreased or increased, respectively, ERalpha mRNA levels. Overexpression of pre-miR-206 reduced ERalpha and beta-actin protein levels, with no effect on ERbeta, E-cadherin, or glyceraldehyde-3-phosphate dehydrogenase. Reporter constructs containing the hERalpha1 or hERalpha2 binding sites inserted into the 3'-untranslated region of the luciferase mRNA conferred a 1.6- and 2.2-fold repression of luciferase activity, respectively, in HeLa cells. Both miR-206 sites responded accordingly to exogenous hsa-pre-miR-206 and 2'-O-methyl antagomiR-206, and both sites were rendered inactive by mutations that disrupted hybridization to the 5'-seed of miR-206. A C-->T single nucleotide polymorphism in the hERalpha1 site increased repression of luciferase activity to approximately 3.3-fold in HeLa cells. MiR-206 levels were higher in ERalpha-negative MB-MDA-231 cells than ERalpha-positive MCF-7 cells, but only the ERalpha1 site mediated significantly more repression in reporter constructs. MiR-206 expression was strongly inhibited by ERalpha agonists, but not by an ERbeta agonist or progesterone, indicating a mutually inhibitory feedback loop. These findings provide the first evidence for the posttranscriptional regulation of ERalpha by a micro-RNA in the context of breast cancer.
Collapse
Affiliation(s)
- Brian D Adams
- Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, Connecticut 06030, USA
| | | | | |
Collapse
|
185
|
Abstract
MicroRNAs (miRNAs) are small RNAs of generally 21 to 23 nt that down-regulate target gene expression at the posttranscriptional level. Although miRNAs are endogenously expressed by all animals and plants, numerous DNA viruses have now been identified that also encode miRNAs, presumably to down-modulate protein expression from viral and host transcripts. Although this has been shown in some cases, the function of the majority of viral miRNAs remains unclear. The herpesviruses stand out by making extensive use of miRNA expression during long-term latent infection or lytic replication. Because viral miRNAs are only present in the context of viral infection and can therefore be considered "exogenous," their expression in uninfected cells of the appropriate cell type is a valuable tool to assess their function. Techniques to achieve and validate the expression of viral miRNAs are described in this review.
Collapse
Affiliation(s)
- Eva Gottwein
- Center for Virology and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | | |
Collapse
|
186
|
Abstract
RNA silencing is a widespread phenomenon that regulates gene expression at different levels. Small RNA molecules are at the core of all RNA silencing pathways and can be grouped in distinct families depending on their chemical properties and their mode of action. Among these small RNAs, microRNAs (miRNAs) represent one of the most extensively studied classes in animals. These tiny endogenous RNAs regulate gene expression posttranscriptionally by cleaving the targeted transcript or by interfering with its translation. miRNAs are found in plants and animals and have been identified in mammalian viruses. This indicates that mammalian viruses exploit the host RNA silencing machinery to produce miRNAs that have the potential to act both on the infected host genome and on the viral genome. The techniques used for identification of viral miRNAs mostly parallel what is used for identifying cellular miRNAs. The use of prediction algorithms followed by validation has been successful. A more direct and nonbiased way of identifying viral miRNAs consists of cloning and sequencing small RNA libraries from virally infected cells or tissues.
Collapse
Affiliation(s)
- Sébastien Pfeffer
- Institut de Biologie Moléculaire des Plantes, CNRS, Strasbourg cedex, France
| |
Collapse
|
187
|
Abstract
Given the important function of microRNAs (miRNAs) in the control of gene expression, it is not surprising to find that some viruses encode their own miRNAs. Although the function of the overwhelming majority of these miRNAs remains unknown, at least some of them are expected to play crucial roles in the viral life cycle, and hence there is great interest in identifying novel viral miRNAs. The majority of currently known viral (and host) miRNAs have been identified by cloning of small RNAs, but, due to their small size, viral genomes are especially amenable to alternative methods based on the computational prediction of miRNA candidates. Here, we provide a detailed protocol on how to use computational prediction methods in conjunction with high-throughput microarray analysis to detect miRNAs in viral genomes.
Collapse
Affiliation(s)
- Christopher S Sullivan
- Department of Molecular Genetics and Microbiology, University of Texas at Austin, Austin, Texas, USA
| | | |
Collapse
|
188
|
Yeung ML, Bennasser Y, Le SY, Jeang KT. RNA interference and HIV-1. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2007; 55:427-38. [PMID: 17586323 DOI: 10.1016/s1054-3589(07)55013-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Man Lung Yeung
- Molecular Virology Section, Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
189
|
Abstract
MicroRNAs (miRNAs) consist of a growing class of non-coding RNAs (ncRNAs) that negatively regulate the expression of genes involved in development, differentiation, proliferation, apoptosis and other important cellular processes. miRNAs are usually 18-25 nt long and are each able to regulate several mRNAs by mechanisms such as incomplete base pairing and Post-Transcriptional Gene Silencing (PTGS). A growing number of reports have shown that aberrant miRNA expression is a common feature of human diseases including cancer, which has sparked interest in targeting these regulators of gene expression as a means of ameliorating these diseases. Here, we review important aspects of miRNA function in normal and pathological states and discuss new modalities of epigenetic intervention strategies that could be used to amend defects in miRNA/mRNA interactions.
Collapse
Affiliation(s)
- T Wurdinger
- Molecular Neurogenetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston/Charlestown, MA, USA.
| | | |
Collapse
|
190
|
Hamza MA, Higgins DM, Ruyechan WT. Herpes simplex virus type-1 latency-associated transcript-induced immunoreactivity of substance P in trigeminal neurons is reversed by bone morphogenetic protein-7. Neurosci Lett 2006; 413:31-5. [PMID: 17239535 PMCID: PMC1821347 DOI: 10.1016/j.neulet.2006.11.063] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Revised: 10/10/2006] [Accepted: 11/07/2006] [Indexed: 11/23/2022]
Abstract
Herpes simplex virus type-1 (HSV-1) primarily infects mucoepithelial tissues of the eye and the orofacial region. Subsequently, the virus is retrogradely transported through the axons of the trigeminal sensory neurons to their nuclei, where the virus establishes a life-long latent infection. During this latency period, the viral genome is transcriptionally silent except for a single region encoding the latency-associated transcript (LAT). To understand how HSV-1 latency might affect the expression of substance P in sensory neurons, we transfected primary cultures of trigeminal neurons obtained from rat embryos, with LAT expressing plasmids. The expression of LAT increased the percentage of substance P-immunoreactive neurons by two thirds. To examine the effect of bone morphogenetic protein-7 (BMP7) on the LAT-induced increase in substance P expression in trigeminal neurons, cultures transfected with LAT were treated with BMP7. Treatment with BMP7 reversed the effects of LAT on substance P expression in trigeminal neurons. Our data show for the first time that LAT increases substance P expression in trigeminal neurons and BMP7 can reverse these effects of LAT.
Collapse
Affiliation(s)
- Mohamed A Hamza
- Departments of Pharmacology and Toxicology, University at Buffalo, SUNY, Buffalo, NY 14214, United States
| | | | | |
Collapse
|
191
|
Moissiard G, Voinnet O. RNA silencing of host transcripts by cauliflower mosaic virus requires coordinated action of the four Arabidopsis Dicer-like proteins. Proc Natl Acad Sci U S A 2006; 103:19593-8. [PMID: 17164336 PMCID: PMC1698440 DOI: 10.1073/pnas.0604627103] [Citation(s) in RCA: 199] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
RNA silencing is an ancient mechanism of gene regulation with important antiviral roles in plants and insects. Although induction of RNA silencing by RNA viruses has been well documented in plants, the interactions between DNA viruses and the host silencing machinery remain poorly understood. We investigate this question with cauliflower mosaic virus (CaMV), a dsDNA virus that expresses its genome through the polycistronic 35S RNA, which carries an unusually extensive secondary structure known as translational leader. We show that CaMV-derived siRNAs accumulate in turnip- and Arabidopsis-infected plants and that the leader is a major, albeit not exclusive, source for those molecules. Biogenesis of leader-derived siRNA requires the coordinated and hierarchical action of the four Arabidopsis Dicer-like (DCL) proteins. Our study also uncovers a "facilitating" role exerted by the microRNA biosynthetic enzyme DCL1 on accumulation of DCL2-, DCL3-, and DCL4-dependent siRNAs derived from the 35S leader. This feature of DCL1 defines a small RNA biosynthetic pathway that might have relevance for endogenous gene regulation. Several leader-derived siRNAs were found to bear near-perfect sequence complementarity to Arabidopsis transcripts, and, using a sensor transgene, we provide direct evidence that at least one of those molecules acts as a bona fide siRNA in infected turnip. Extensive bioinformatics searches identified >100 transcripts potentially targeted by CaMV-derived siRNAs, several of which are effectively down-regulated during infection. The implications of virus-directed silencing of host gene expression are discussed.
Collapse
Affiliation(s)
- Guillaume Moissiard
- Institut de Biologie Moléculaire des Plantes, Centre National de la Recherche Scientifique, Unité Propre de Recherche 2357, 12 Rue du Général Zimmer, 67084 Strasbourg Cedex, France
| | - Olivier Voinnet
- Institut de Biologie Moléculaire des Plantes, Centre National de la Recherche Scientifique, Unité Propre de Recherche 2357, 12 Rue du Général Zimmer, 67084 Strasbourg Cedex, France
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
192
|
Ushijima Y, Luo C, Goshima F, Yamauchi Y, Kimura H, Nishiyama Y. Determination and analysis of the DNA sequence of highly attenuated herpes simplex virus type 1 mutant HF10, a potential oncolytic virus. Microbes Infect 2006; 9:142-9. [PMID: 17218138 DOI: 10.1016/j.micinf.2006.10.019] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 10/29/2006] [Accepted: 10/31/2006] [Indexed: 02/04/2023]
Abstract
A spontaneously occurring herpes simplex virus type 1 (HSV-1) mutant, designated HF10, replicates very efficiently and induces extensive cell fusion in most transformed cells as well as Vero cells, but is highly attenuated in mice when inoculated by peripheral routes of infection. Recent studies have shown that HF10 is a promising agent for use in oncolytic virotherapy. In this study, we sequenced the genome of HF10 and compared it with that of HSV-1 strain 17, a reference strain with the syn+ phenotype. The sequencing covered whole regions corresponding to all open reading frames of strain 17, and the overall putative amino acid identity between HF10 and strain 17 was 99.1% except for proteins encoded by three genes with frame-shift mutations. HF10 had a number of deletions and insertions in the genome, resulting in the lack of the functional expression of UL43, UL49.5, UL55, UL56 and latency-associated transcripts. Additionally, HF10 had amino acid changes in genes involved in the regulation of syncytium formation, including UL1, UL20, UL22, UL24, UL27 and UL53. The proteins encoded by UL1, UL2, UL11, UL44, US1, US7, US8.5, US10 and US12 exhibited a relatively high divergence. These data provide the genetic background of HF10 and insight into the molecular mechanism of HSV-1 replication and pathogenicity.
Collapse
Affiliation(s)
- Yoko Ushijima
- Department of Virology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | | | | | | | | | |
Collapse
|
193
|
Margolis TP, Imai Y, Yang L, Vallas V, Krause PR. Herpes simplex virus type 2 (HSV-2) establishes latent infection in a different population of ganglionic neurons than HSV-1: role of latency-associated transcripts. J Virol 2006; 81:1872-8. [PMID: 17151134 PMCID: PMC1797553 DOI: 10.1128/jvi.02110-06] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) and HSV-2 cause very similar acute infections but differ in their abilities to reactivate from trigeminal and dorsal root ganglia. To investigate differences in patterns of viral infection, we colabeled murine sensory ganglia for evidence of HSV infection and for the sensory neuron marker A5 or KH10. During acute infection, 7 to 10% of HSV-1 or HSV-2 antigen-positive neurons were A5 positive and 13 to 16% were KH10 positive, suggesting that both viruses reach each type of neuron in a manner proportional to their representation in uninfected ganglia. In murine trigeminal ganglia harvested during HSV latency, 25% of HSV-1 latency-associated transcript (LAT)- and 4% of HSV-2 LAT-expressing neurons were A5 positive, while 12% of HSV-1 LAT- and 42% of HSV-2 LAT-expressing neurons were KH10 positive. A similar difference was observed in murine dorsal root ganglia. These differences could not be attributed to differences in LAT expression levels in A5- versus KH10-positive neurons. Thus, HSV-1 demonstrated a preference for the establishment of latency in A5-positive neurons, while HSV-2 demonstrated a preference for the establishment of latency in KH10-positive neurons. A chimeric HSV-2 mutant that expresses the HSV-1 LAT exhibited an HSV-1 phenotype, preferentially establishing latency in A5-positive neurons. These data imply that the HSV-1 and HSV-2 LAT regions influence the ability of virus to establish latency in different neuronal subtypes. That the same chimeric virus has a characteristic HSV-1 reactivation phenotype further suggests that LAT-influenced establishment of latency in specific neuronal subtypes could be an important part of the mechanism by which LAT influences viral reactivation phenotypes.
Collapse
Affiliation(s)
- Todd P Margolis
- F. I. Proctor Foundation, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | | | | | | | |
Collapse
|
194
|
Sciortino MT, Perri D, Medici MA, Grelli S, Serafino A, Borner C, Mastino A. Role of Bcl-2 expression for productive herpes simplex virus 2 replication. Virology 2006; 356:136-146. [PMID: 16950491 DOI: 10.1016/j.virol.2006.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Revised: 06/22/2006] [Accepted: 08/02/2006] [Indexed: 11/20/2022]
Abstract
Herpes simplex viruses infect a variety of cells in vitro. However, not all infected cells sustain a fully productive replication of these viruses. We have shown that, in U937 monocytoid cells, herpes simplex virus 2 (HSV-2) causes a low-productive infection characterized by apoptosis as cytopathic effect at a late stage of infection. This effect was associated with a down-regulation of the Bcl-2 protein. We therefore asked whether destabilization of Bcl-2 expression could act as a limiting factor for the productive HSV-2 infection. We found that overexpression of Bcl-2 in U937 cells dramatically increased the capability of these cells to sustain a fully productive infection, while protecting against apoptosis induced by HSV-2. Overall, our data indicate that Bcl-2 expression acts as a regulator of HSV-2 replication.
Collapse
Affiliation(s)
- Maria Teresa Sciortino
- Department of Microbiological, Genetic and Molecular Sciences, University of Messina, Salita Sperone 31, 98166 Messina, Italy
| | | | | | | | | | | | | |
Collapse
|
195
|
Kulesza CA, Shenk T. Murine cytomegalovirus encodes a stable intron that facilitates persistent replication in the mouse. Proc Natl Acad Sci U S A 2006; 103:18302-7. [PMID: 17105807 PMCID: PMC1838746 DOI: 10.1073/pnas.0608718103] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The human cytomegalovirus immediate-early 5-kb RNA previously has been shown to be a stable intron that is not required for efficient replication of the virus in cultured fibroblasts. Here we describe a murine cytomegalovirus 7.2-kb ortholog of the human cytomegalovirus 5-kb RNA. The 5' end of the 7.2-kb transcript maps to a consensus splice-donor site that is conserved among all cytomegaloviruses. We constructed mutant viruses lacking the entire 7.2-kb coding domain, the splice-donor site predicted to function in the generation of the intron or a hairpin predicted to stabilize the intron. All three mutant viruses failed to produce the 7.2-kb RNA, supporting our conclusion that it is a stable intron. Each of the mutants replicated with normal kinetics in cultured fibroblasts, but the mutants exhibited a clear defect within infected mice. Although the initial acute phase at 4 days after infection appeared to be normal, none of the mutant viruses progressed to the persistent phase, i.e., little virus was detected in the salivary gland at 14 days after infection. The intron functions as an in vivo virulence factor, facilitating progression from the acute to persistent phase of infection.
Collapse
Affiliation(s)
| | - Thomas Shenk
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
196
|
Hüfner K, Derfuss T, Herberger S, Sunami K, Russell S, Sinicina I, Arbusow V, Strupp M, Brandt T, Theil D. Latency of alpha-herpes viruses is accompanied by a chronic inflammation in human trigeminal ganglia but not in dorsal root ganglia. J Neuropathol Exp Neurol 2006; 65:1022-30. [PMID: 17021407 DOI: 10.1097/01.jnen.0000235852.92963.bf] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The immune response to latent herpesvirus infections was compared in human trigeminal ganglia (TG) and dorsal root ganglia (DRG) of 15 dead individuals. On the basis of our previous findings, we hypothesized that T-cells would be attracted to sensory neurons latently infected with herpes simplex virus type 1 (HSV-1), but not to those harboring latent varicella zoster virus (VZV). We showed that the TG contain a positive hybridization signal for HSV-1 latency-associated transcript (LAT), whereas the DRG from the same individuals lack detectable LAT. In contrast, immunohistochemistry revealed that latent VZV protein 62 stained positive in the vast majority of all tested TG and DRG. T-cell infiltrates prominently surrounded individual neurons in the TG but not in the DRG. TaqMan polymerase chain reaction also showed higher expression of CD8 and RANTES transcripts in the TG versus DRG. Only the infiltrates in the TG, but not in the DRG, produced RANTES at the protein level. Because it has been shown that RANTES protein is produced only after T-cell receptor stimulation, we assume that T-cell infiltration is associated with antigen recognition in the TG but not in the DRG.
Collapse
Affiliation(s)
- Katharina Hüfner
- Department of Neurology, Klinikum Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs of about 18-24 nucleotides in length that negatively regulate gene expression. Discovered only recently, it has become clear that they are involved in many biological processes such as developmental timing, differentiation and cell death. Data that connect miRNAs to various kinds of diseases, particularly cancer, are accumulating. miRNAs can influence cancer development in many ways, including the regulation of cell proliferation, cell transformation, and cell death. In this review, we focus on miRNAs that have been shown to play a role in the regulation of apoptosis. We first describe in detail how Drosophila has been utilized as a model organism to connect several miRNAs with the cell death machinery. We discuss the genetic approaches that led to the identification of those miRNAs and subsequent work that helped to establish their function. In the second part of the review article, we focus on the involvement of miRNAs in apoptosis regulation in mammals. Intriguingly, many of the miRNAs that regulate apoptosis have been shown to affect cancer development. In the end, we discuss a virally encoded miRNA that influences the cell death response in the mammalian host cell. In summary, the data gathered over the recent years clearly show the potential and important role of miRNAs to regulate apoptosis at various levels and in several organisms.
Collapse
Affiliation(s)
- M Jovanovic
- Institute of Molecular Biology, University of Zurich and ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
198
|
Abstract
Viruses represent one of the main factors that cause normal cells to proliferate and to become malignant: up to 15% of all human cancers are associated with single or multiple virus infections, and several viruses have been recognized as causal agents of specific types of cancer. Viruses have evolved many strategies to prevent infected cells from becoming apoptotic and to evade the innate and adaptive immune responses of their hosts. The recent discovery that Epstein-Barr virus and other herpesviruses produce their own sets of micro (mi)RNAs brings an additional layer of complexity in this ongoing host-virus arms race and changes our initial views of the antiviral roles of RNA silencing in plants and insects. It seems that, rather than being inhibited by this process, many mammalian viruses can usurp or divert the host RNA silencing machinery to their advantage. Viral-encoded miRNAs can act both in cis, to ensure accurate expression of viral genomes, and in trans, to modify the expression of host transcripts. Here, we review the current knowledge on viral miRNAs and discuss how mammalian viruses can also perturb host miRNA expression. Those recent findings provide new insights into the role of viruses and miRNAs in cancer development.
Collapse
Affiliation(s)
- S Pfeffer
- IBMP-CNRS, rue du Général Zimmer, Strasbourg, France.
| | | |
Collapse
|
199
|
Hamza MA, Higgins DM, Ruyechan WT. Herpes simplex virus type-1 latency inhibits dendritic growth in sympathetic neurons. Neurobiol Dis 2006; 24:367-73. [PMID: 16952455 DOI: 10.1016/j.nbd.2006.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Revised: 07/17/2006] [Accepted: 07/19/2006] [Indexed: 11/21/2022] Open
Abstract
Herpes simplex virus type-1 (HSV-1) initially infects mucoepithelial tissues of the orofacial region, the eye and to a lesser extent the genitalia. Subsequently, the virus is retrogradely transported through the axons of the sensory and sympathetic neurons to their nuclei, where the virus establishes a life-long latent infection. During this latency period, the viral genome is transcriptionally silent except for a single region encoding the latency-associated transcript (LAT). LAT has been shown to affect apoptosis, but little else is known regarding its effects on neurons. To understand how HSV-1 latency might affect dendrites in sympathetic neurons, we transfected primary cultures of sympathetic neurons obtained from rat embryos, with LAT expressing plasmids. LAT inhibited initial dendritic growth and induced dendritic retraction in sympathetic neurons. Latent HSV-1 infection of cultured sympathetic neurons inhibited dendritic growth indicating that this is likely also a consequence of natural infection.
Collapse
MESH Headings
- Animals
- Bone Morphogenetic Protein 7
- Bone Morphogenetic Proteins/genetics
- Bone Morphogenetic Proteins/metabolism
- Cell Differentiation/genetics
- Cells, Cultured
- Dendrites/pathology
- Dendrites/virology
- Encephalitis, Herpes Simplex/genetics
- Encephalitis, Herpes Simplex/metabolism
- Encephalitis, Herpes Simplex/physiopathology
- Ganglia, Sympathetic/growth & development
- Ganglia, Sympathetic/physiopathology
- Ganglia, Sympathetic/virology
- Gene Expression Regulation, Viral/genetics
- Genetic Vectors/genetics
- Green Fluorescent Proteins/genetics
- Herpes Simplex/genetics
- Herpes Simplex/metabolism
- Herpes Simplex/physiopathology
- Herpesvirus 1, Human/physiology
- MicroRNAs
- Rats
- Rats, Sprague-Dawley
- Signal Transduction/genetics
- Transfection
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Viral Proteins/genetics
- Virus Latency/physiology
Collapse
Affiliation(s)
- Mohamed A Hamza
- Department of Pharmacology and Toxicology, University at Buffalo, SUNY, Buffalo, NY 14214, USA
| | | | | |
Collapse
|
200
|
Han JY, Sloan DD, Aubert M, Miller SA, Dang CH, Jerome KR. Apoptosis and antigen receptor function in T and B cells following exposure to herpes simplex virus. Virology 2006; 359:253-63. [PMID: 17067652 PMCID: PMC1868478 DOI: 10.1016/j.virol.2006.09.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Revised: 06/21/2006] [Accepted: 09/22/2006] [Indexed: 11/17/2022]
Abstract
T cells are an essential component of the immune response against herpes simplex virus (HSV) infection. We previously reported that incubation of T cells with HSV-infected fibroblasts inhibits subsequent T cell antigen receptor signal transduction. In the current study, we found that incubation of T cells with HSV-infected fibroblasts also leads to apoptosis in exposed T cells. Apoptosis was observed in Jurkat cells, a T cell leukemia line, and also in CD4(+) cells isolated from human peripheral blood mononuclear cells. Direct infection of these cells with HSV also resulted in apoptosis. Clinical isolates of both HSV type 1 and 2 induced apoptosis in infected T cells at comparable levels to cells infected with laboratory strains of HSV, suggesting an immune evasion mechanism that may be clinically relevant. Further understanding of these viral immune evasion mechanisms could be exploited for better management of HSV infection.
Collapse
Affiliation(s)
- Jin-Young Han
- Department of Pediatrics, University of Washington, Seattle, WA 98195
- Program in Infectious Diseases, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
- Division of Infectious Diseases, Immunology and Rheumatology, Children’s Hospital and Regional Medical Center, Seattle, WA 98105
| | - Derek D. Sloan
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195
- Program in Infectious Diseases, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Martine Aubert
- Program in Infectious Diseases, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Sara A. Miller
- Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195
| | - Chung H. Dang
- Program in Infectious Diseases, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Keith R. Jerome
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195
- Department of Microbiology, University of Washington, Seattle, WA 98195
- Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195
- Program in Infectious Diseases, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
- *Corresponding author: Keith R. Jerome, M.D., Ph.D., Program in Infectious Diseases, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, D3-100, Seattle, WA, 98109. E-mail address:
| |
Collapse
|