1
|
Kawasaki H, Hariyama T, Kosugi I, Meguro S, Iwata F, Shimizu K, Magata Y, Iwashita T. Human induced pluripotent stem cells are resistant to human cytomegalovirus infection primarily at the attachment level due to the reduced expression of cell-surface heparan sulfate. J Virol 2024; 98:e0127823. [PMID: 38345384 PMCID: PMC10949504 DOI: 10.1128/jvi.01278-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/16/2024] [Indexed: 03/20/2024] Open
Abstract
Cytomegalovirus (CMV), a type of herpes virus, is the predominant cause of congenital anomalies due to intrauterine infections in humans. Adverse outcomes related to intrauterine infections with human cytomegalovirus (HCMV) vary widely, depending on factors such as fetal infection timing, infection route, and viral virulence. The precise mechanism underlying HCMV susceptibility remains unclear. In this study, we compared the susceptibility of neonatal human dermal fibroblast cells (NHDFCs) and human induced pluripotent stem cells (hiPSCs) derived from NHDFCs, which are genetically identical to HCMV, using immunostaining, microarray, in situ hybridization, quantitative PCR, and scanning electron microscopy. These cells were previously used to compare CMV susceptibility, but the underlying mechanisms were not fully elucidated. HCMV susceptibility of hiPSCs was significantly lower in the earliest phase. No shared gene ontologies were observed immediately post-infection between the two cell types using microarray analysis. Early-stage expression of HCMV antigens and the HCMV genome was minimal in immunostaining and in in situ hybridization in hiPSCs. This strongly suggests that HCMV does not readily bind to hiPSC surfaces. Scanning electron microscopy performed using the NanoSuit method confirmed the scarcity of HCMV particles on hiPSC surfaces. The zeta potential and charge mapping of the charged surface in NHDFCs and hiPSCs exhibited minimal differences when assessed using zeta potential analyzer and scanning ion conductance microscopy; however, the expression of heparan sulfate (HS) was significantly lower in hiPSCs compared with that in NHDFCs. Thus, HS expression could be a primary determinant of HCMV resistance in hiPSCs at the attachment level. IMPORTANCE Numerous factors such as attachment, virus particle entry, transcription, and virus particle egress can affect viral susceptibility. Since 1984, pluripotent cells are known to be CMV resistant; however, the exact mechanism underlying this resistance remains elusive. Some researchers suggest inhibition in the initial phase of HCMV binding, while others have suggested the possibility of a sufficient amount of HCMV entering the cells to establish latency. This study demonstrates that HCMV particles rarely attach to the surfaces of hiPSCs. This is not due to limitations in the electrostatic interactions between the surface of hiPSCs and HCMV particles, but due to HS expression. Therefore, HS expression should be recognized as a key factor in determining the susceptibility of HCMV in congenital infection in vitro and in vivo. In the future, drugs targeting HS may become crucial for the treatment of congenital CMV infections. Thus, further research in this area is warranted.
Collapse
Affiliation(s)
- Hideya Kawasaki
- Institute for NanoSuit Research, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Takahiko Hariyama
- Institute for NanoSuit Research, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Isao Kosugi
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Shiori Meguro
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Futoshi Iwata
- Research Institute of Electronics, Shizuoka University, Hamamatsu, Shizuoka, Japan
| | - Kosuke Shimizu
- Department of Molecular Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Yasuhiro Magata
- Department of Molecular Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Toshihide Iwashita
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
2
|
CD34 + Hematopoietic Progenitor Cell Subsets Exhibit Differential Ability To Maintain Human Cytomegalovirus Latency and Persistence. J Virol 2021; 95:JVI.02105-20. [PMID: 33177198 DOI: 10.1128/jvi.02105-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 11/20/2022] Open
Abstract
In human cytomegalovirus (HCMV)-seropositive patients, CD34+ hematopoietic progenitor cells (HPCs) provide an important source of latent virus that reactivates following cellular differentiation into tissue macrophages. Multiple groups have used primary CD34+ HPCs to investigate mechanisms of viral latency. However, analyses of mechanisms of HCMV latency have been hampered by the genetic variability of CD34+ HPCs from different donors, availability of cells, and low frequency of reactivation. In addition, multiple progenitor cell types express surface CD34, and the frequencies of these populations differ depending on the tissue source of the cells and culture conditions in vitro In this study, we generated CD34+ progenitor cells from two different embryonic stem cell (ESC) lines, WA01 and WA09, to circumvent limitations associated with primary CD34+ HPCs. HCMV infection of CD34+ HPCs derived from either WA01 or WA09 ESCs supported HCMV latency and induced myelosuppression similar to infection of primary CD34+ HPCs. Analysis of HCMV-infected primary or ESC-derived CD34+ HPC subpopulations indicated that HCMV was able to establish latency and reactivate in CD38+ CD90+ and CD38+/low CD90- HPCs but persistently infected CD38- CD90+ cells to produce infectious virus. These results indicate that ESC-derived CD34+ HPCs can be used as a model for HCMV latency and that the virus either latently or persistently infects specific subpopulations of CD34+ cells.IMPORTANCE Human cytomegalovirus infection is associated with severe disease in transplant patients and understanding how latency and reactivation occur in stem cell populations is essential to understand disease. CD34+ hematopoietic progenitor cells (HPCs) are a critical viral reservoir; however, these cells are a heterogeneous pool with donor-to-donor variation in functional, genetic, and phenotypic characteristics. We generated a novel system using embryonic stem cell lines to model HCMV latency and reactivation in HPCs with a consistent cellular background. Our study defined three key stem cell subsets with differentially regulated latent and replicative states, which provide cellular candidates for isolation and treatment of transplant-mediated disease. This work provides a direction toward developing strategies to control the switch between latency and reactivation.
Collapse
|
3
|
Regulation of the MIE Locus During HCMV Latency and Reactivation. Pathogens 2020; 9:pathogens9110869. [PMID: 33113934 PMCID: PMC7690695 DOI: 10.3390/pathogens9110869] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous herpesviral pathogen that results in life-long infection. HCMV maintains a latent or quiescent infection in hematopoietic cells, which is broadly defined by transcriptional silencing and the absence of de novo virion production. However, upon cell differentiation coupled with immune dysfunction, the virus can reactivate, which leads to lytic replication in a variety of cell and tissue types. One of the mechanisms controlling the balance between latency and reactivation/lytic replication is the regulation of the major immediate-early (MIE) locus. This enhancer/promoter region is complex, and it is regulated by chromatinization and associated factors, as well as a variety of transcription factors. Herein, we discuss these factors and how they influence the MIE locus, which ultimately impacts the phase of HCMV infection.
Collapse
|
4
|
Collins-McMillen D, Buehler J, Peppenelli M, Goodrum F. Molecular Determinants and the Regulation of Human Cytomegalovirus Latency and Reactivation. Viruses 2018; 10:E444. [PMID: 30127257 PMCID: PMC6116278 DOI: 10.3390/v10080444] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a beta herpesvirus that establishes a life-long persistence in the host, like all herpesviruses, by way of a latent infection. During latency, viral genomes are maintained in a quieted state. Virus replication can be reactivated from latency in response to changes in cellular signaling caused by stress or differentiation. The past decade has brought great insights into the molecular basis of HCMV latency. Here, we review the complex persistence of HCMV with consideration of latent reservoirs, viral determinants and their host interactions, and host signaling and the control of cellular and viral gene expression that contributes to the establishment of and reactivation from latency.
Collapse
Affiliation(s)
| | - Jason Buehler
- BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA.
| | | | - Felicia Goodrum
- BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA.
- Department of Immunobiology, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
5
|
Kawasaki H. Pluripotent stem cells are protected from cytomegalovirus infection at multiple points: implications of a new pathogenesis for congenital anomaly caused by cytomegalovirus. Congenit Anom (Kyoto) 2012; 52:147-54. [PMID: 22925215 DOI: 10.1111/j.1741-4520.2012.00375.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In humans, the cytomegalovirus (CMV) is the most significant cause of intrauterine infections that cause congenital anomalies. Intrauterine infection with human CMV is thought to be responsible for a variety of abnormalities, including mental retardation, microcephaly, developmental delay, seizure disorders, and cerebral palsy, depending on the timing of the fetal infection, the infectious route, and the virulence of the virus. In addition to the adaptive immune system, the embryo has potential resistance to CMV during early embryogenesis. Embryonic stem (ES) cells are more resistant to CMV than most other cell types, although the mechanism responsible for this resistance is not well understood. ES cells allow approximately 20-fold less murine CMV (MCMV) DNA to enter the nucleus than mouse embryonic fibroblasts (MEFs), and this inhibition occurs in a multistep manner. In situ hybridization showed that ES cell nuclei had significantly less MCMV DNA than MEF nuclei. This finding appears to be supported by the fact that ES cells express less heparan sulfate, β1-integrin, and vimentin and have fewer nuclear pores than differentiated cells such as MEF. This may reduce the ability of MCMV to attach to and enter the cellular membrane, translocate to the nucleus, and cross the nuclear membrane in pluripotent stem cells (ES-induced pluripotent stem cells). This finding may indicate a new pathogenesis for the congenital anomaly caused by CMV.
Collapse
Affiliation(s)
- Hideya Kawasaki
- Department of Regenerative & Infectious Pathology, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan.
| |
Collapse
|
6
|
Kawasaki H, Kosugi I, Arai Y, Iwashita T, Tsutsui Y. Mouse embryonic stem cells inhibit murine cytomegalovirus infection through a multi-step process. PLoS One 2011; 6:e17492. [PMID: 21407806 PMCID: PMC3047572 DOI: 10.1371/journal.pone.0017492] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 02/07/2011] [Indexed: 01/21/2023] Open
Abstract
In humans, cytomegalovirus (CMV) is the most significant infectious cause of intrauterine infections that cause congenital anomalies of the central nervous system. Currently, it is not known how this process is affected by the timing of infection and the susceptibility of early-gestational-period cells. Embryonic stem (ES) cells are more resistant to CMV than most other cell types, although the mechanism responsible for this resistance is not well understood. Using a plaque assay and evaluation of immediate-early 1 mRNA and protein expression, we found that mouse ES cells were resistant to murine CMV (MCMV) at the point of transcription. In ES cells infected with MCMV, treatment with forskolin and trichostatin A did not confer full permissiveness to MCMV. In ES cultures infected with elongation factor-1α (EF-1α) promoter-green fluorescent protein (GFP) recombinant MCMV at a multiplicity of infection of 10, less than 5% of cells were GFP-positive, despite the fact that ES cells have relatively high EF-1α promoter activity. Quantitative PCR analysis of the MCMV genome showed that ES cells allow approximately 20-fold less MCMV DNA to enter the nucleus than mouse embryonic fibroblasts (MEFs) do, and that this inhibition occurs in a multi-step manner. In situ hybridization revealed that ES cell nuclei have significantly less MCMV DNA than MEF nuclei. This appears to be facilitated by the fact that ES cells express less heparan sulfate, β1 integrin, and vimentin, and have fewer nuclear pores, than MEF. This may reduce the ability of MCMV to attach to and enter through the cellular membrane, translocate to the nucleus, and cross the nuclear membrane in pluripotent stem cells (ES/induced pluripotent stem cells). The results presented here provide perspective on the relationship between CMV susceptibility and cell differentiation.
Collapse
Affiliation(s)
- Hideya Kawasaki
- Department of Second Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | | | | | | | | |
Collapse
|
7
|
Zydek M, Hagemeier C, Wiebusch L. Cyclin-dependent kinase activity controls the onset of the HCMV lytic cycle. PLoS Pathog 2010; 6:e1001096. [PMID: 20844576 PMCID: PMC2936547 DOI: 10.1371/journal.ppat.1001096] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 08/11/2010] [Indexed: 12/21/2022] Open
Abstract
The onset of human cytomegalovirus (HCMV) lytic infection is strictly synchronized with the host cell cycle. Infected G0/G1 cells support viral immediate early (IE) gene expression and proceed to the G1/S boundary where they finally arrest. In contrast, S/G2 cells can be infected but effectively block IE gene expression and this inhibition is not relieved until host cells have divided and reentered G1. During latent infection IE gene expression is also inhibited, and for reactivation to occur this block to IE gene expression must be overcome. It is only poorly understood which viral and/or cellular activities maintain the block to cell cycle or latency-associated viral IE gene repression and whether the two mechanisms may be linked. Here, we show that the block to IE gene expression during S and G2 phase can be overcome by both genotoxic stress and chemical inhibitors of cellular DNA replication, pointing to the involvement of checkpoint-dependent signaling pathways in controlling IE gene repression. Checkpoint-dependent rescue of IE expression strictly requires p53 and in the absence of checkpoint activation is mimicked by proteasomal inhibition in a p53 dependent manner. Requirement for the cyclin dependent kinase (CDK) inhibitor p21 downstream of p53 suggests a pivotal role for CDKs in controlling IE gene repression in S/G2 and treatment of S/G2 cells with the CDK inhibitor roscovitine alleviates IE repression independently of p53. Importantly, CDK inhibiton also overcomes the block to IE expression during quiescent infection of NTera2 (NT2) cells. Thus, a timely block to CDK activity not only secures phase specificity of the cell cycle dependent HCMV IE gene expression program, but in addition plays a hitherto unrecognized role in preventing the establishment of a latent-like state.
Collapse
Affiliation(s)
- Martin Zydek
- Children's Hospital, Laboratory for Molecular Biology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Free University of Berlin, Faculty of Biology, Chemistry and Pharmacy, Berlin, Germany
| | - Christian Hagemeier
- Children's Hospital, Laboratory for Molecular Biology, Charité Universitätsmedizin Berlin, Berlin, Germany
- * E-mail: (LW); (CH)
| | - Lüder Wiebusch
- Children's Hospital, Laboratory for Molecular Biology, Charité Universitätsmedizin Berlin, Berlin, Germany
- * E-mail: (LW); (CH)
| |
Collapse
|
8
|
Properties of virion transactivator proteins encoded by primate cytomegaloviruses. Virol J 2009; 6:65. [PMID: 19473490 PMCID: PMC2693105 DOI: 10.1186/1743-422x-6-65] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Accepted: 05/27/2009] [Indexed: 11/25/2022] Open
Abstract
Background Human cytomegalovirus (HCMV) is a betaherpesvirus that causes severe disease in situations where the immune system is immature or compromised. HCMV immediate early (IE) gene expression is stimulated by the virion phosphoprotein pp71, encoded by open reading frame (ORF) UL82, and this transactivation activity is important for the efficient initiation of viral replication. It is currently recognized that pp71 acts to overcome cellular intrinsic defences that otherwise block viral IE gene expression, and that interactions of pp71 with the cell proteins Daxx and ATRX are important for this function. A further property of pp71 is the ability to enable prolonged gene expression from quiescent herpes simplex virus type 1 (HSV-1) genomes. Non-human primate cytomegaloviruses encode homologs of pp71, but there is currently no published information that addresses their effects on gene expression and modes of action. Results The UL82 homolog encoded by simian cytomegalovirus (SCMV), strain Colburn, was identified and cloned. This ORF, named S82, was cloned into an HSV-1 vector, as were those from baboon, rhesus monkey and chimpanzee cytomegaloviruses. The use of an HSV-1 vector enabled expression of the UL82 homologs in a range of cell types, and permitted investigation of their abilities to direct prolonged gene expression from quiescent genomes. The results show that all UL82 homologs activate gene expression, and that neither host cell type nor promoter target sequence has major effects on these activities. Surprisingly, the UL82 proteins specified by non-human primate cytomegaloviruses, unlike pp71, did not direct long term expression from quiescent HSV-1 genomes. In addition, significant differences were observed in the intranuclear localization of the UL82 homologs, and in their effects on Daxx. Strikingly, S82 mediated the release of Daxx from nuclear domain 10 substructures much more rapidly than pp71 or the other proteins tested. All UL82 homologs stimulated the early release of ATRX from nuclear domain 10. Conclusion All of the UL82 homolog proteins analysed activated gene expression, but surprising differences in other aspects of their properties were revealed. The results provide new information on early events in infection with cytomegaloviruses.
Collapse
|
9
|
Neuropathogenesis of congenital cytomegalovirus infection: disease mechanisms and prospects for intervention. Clin Microbiol Rev 2009; 22:99-126, Table of Contents. [PMID: 19136436 DOI: 10.1128/cmr.00023-08] [Citation(s) in RCA: 324] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Congenital cytomegalovirus (CMV) infection is the leading infectious cause of mental retardation and hearing loss in the developed world. In recent years, there has been an improved understanding of the epidemiology, pathogenesis, and long-term disabilities associated with CMV infection. In this review, current concepts regarding the pathogenesis of neurological injury caused by CMV infections acquired by the developing fetus are summarized. The pathogenesis of CMV-induced disabilities is considered in the context of the epidemiology of CMV infection in pregnant women and newborn infants, and the clinical manifestations of brain injury are reviewed. The prospects for intervention, including antiviral therapies and vaccines, are summarized. Priorities for future research are suggested to improve the understanding of this common and disabling illness of infancy.
Collapse
|
10
|
Kitagawa R, Takahashi Y, Takahashi M, Imazu H, Yasuda M, Sadanari H, Tanaka J. Hexamethylene bisacetamide can convert nonpermissive human cells to a permissive state for expressing the major immediate-early genes of human cytomegalovirus by up-regulating NF-kappaB activity. Virology 2008; 383:195-206. [PMID: 19027925 DOI: 10.1016/j.virol.2008.10.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Revised: 08/05/2008] [Accepted: 10/17/2008] [Indexed: 11/26/2022]
Abstract
Expression of the major immediate-early (MIE) genes of human cytomegalovirus (HCMV) in the human thyroid papillary carcinoma cell line TPC-1 is repressed at the transcriptional level. However, treatment of these cells with hexamethylene bisacetamide (HMBA), a chemical inducer of differentiation, for 12 to 24 h before infection enabled the cells to support IE1 and IE2 gene expression and consequently HCMV replication. In HMBA-treated cells the transcription factor NF-kappaB was induced and the MIE promoter (MIEP) was activated. The presence of a NF-kappaB inhibitory peptide SN-50 or expression of a dominant negative IkappaBalpha protein during the HMBA pretreatment period efficiently prevented the HMBA-induced MIEP activation and MIE protein synthesis. Moreover, introduction of mutations into the NF-kappaB binding sites in the MIEP in a plasmid expressing the IE1 protein diminished its ability to express the protein in HMBA-treated cells. Therefore, the NF-kappaB activity previously induced in HMBA-treated cells and the NF-kappaB sites in the MIEP were shown to be essential for HCMV to respond to HMBA action and to express the MIE genes. Investigation of the mechanisms by which HMBA activates NF-kappaB revealed that degradation of IkappaBalpha and translocation of the phosphorylated NF-kappaB p65 subunit to the nucleus, both of which are known to be critical steps in NF-kappaB activation, are stimulated in the HMBA-treated cells. These results indicate that treatment of nonpermissive TPC-1 cells with HMBA induces MIE gene permissiveness by up-regulating NF-kappaB activity.
Collapse
Affiliation(s)
- Ryoko Kitagawa
- Department of Clinical Laboratory Science, School of Health Sciences, College of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kodatsuno, Ishikawa, Japan
| | | | | | | | | | | | | |
Collapse
|
11
|
Luo J, Cun W, Che Y, Wang L, Li W, Liu L, Li Q. Analysis of HSV-I ICP22 effects on HCMV major immediate-early promoter structure. ACTA ACUST UNITED AC 2007; 50:292-7. [PMID: 17609884 DOI: 10.1007/s11427-007-0041-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Accepted: 11/22/2006] [Indexed: 10/23/2022]
Abstract
The human cytomegalovirus (HCMV) major immediate-early (MIE) promoter has strong transcriptional promoting capability. Its cis-acting regulatory elements form a special structure in this region that is repeated multiple times; the biological significance of these elements and their different compositions in the transcriptional promoting process remain unclear. Our results demonstrate that the HSV-I MIE protein ICP22 can generate strong repression of many viral and cellular promoters and enhancers. We further studied the transcriptional effects of ICP22 on structural elements and mutations in various HCMV MIE promoters by using a CAT assay. In spite of different transcriptional effects of all the elements in the presence of ICP22, the transcriptional efficiencies exhibited by mutations generated by different compositions and an entire HCMV promoter, are not the simple sum of the functions of these elements. Furthermore, the transcriptional activities of specific sequences were not affected by the presence of ICP22. Therefore, it is assumed that the HCMV MIE promoter co-regulates expression of downstream genes by using viral and cellular specific factors via a specific pathway.
Collapse
Affiliation(s)
- Jie Luo
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650118, China
| | | | | | | | | | | | | |
Collapse
|
12
|
Saffert RT, Kalejta RF. Human cytomegalovirus gene expression is silenced by Daxx-mediated intrinsic immune defense in model latent infections established in vitro. J Virol 2007; 81:9109-20. [PMID: 17596307 PMCID: PMC1951389 DOI: 10.1128/jvi.00827-07] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In addition to productive lytic infections, herpesviruses such as human cytomegalovirus (HCMV) establish a reservoir of latently infected cells that permit lifelong colonization of the host. When latency is established, the viral immediate-early (IE) genes that initiate the lytic replication cycle are not expressed. HCMV IE gene expression at the start of a lytic infection is facilitated by the viral pp71 protein, which is delivered to cells by infectious viral particles. pp71 neutralizes the Daxx-mediated cellular intrinsic immune defense that silences IE gene expression by generating a repressive chromatin structure on the viral major IE promoter (MIEP). In naturally latently infected cells and in cells latently infected in vitro, the MIEP also adopts a similar silenced chromatin structure. Here we analyze the role of Daxx in quiescent HCMV infections in vitro that mimic some, but not all, of the characteristics of natural latency. We show that in these "latent-like" infections, the Daxx-mediated defense that represses viral gene expression is not disabled because pp71 and Daxx localize to different cellular compartments. We demonstrate that Daxx is required to establish quiescent HCMV infections in vitro because in cells that would normally foster the establishment of these latent-like infections, the loss of Daxx causes the lytic replication cycle to be initiated. Importantly, the lytic cycle is inefficiently completed, which results in an abortive infection. Our work demonstrates that, in certain cell types, HCMV must silence its own gene expression to establish quiescence and prevent abortive infection and that the virus usurps a Daxx-mediated cellular intrinsic immune defense mechanism to do so. This identifies Daxx as one of the likely multiple viral and cellular determinants in the pathway of HCMV quiescence in vitro, and perhaps in natural latent infections as well.
Collapse
Affiliation(s)
- Ryan T Saffert
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | |
Collapse
|
13
|
Keller MJ, Wu AW, Andrews JI, McGonagill PW, Tibesar EE, Meier JL. Reversal of human cytomegalovirus major immediate-early enhancer/promoter silencing in quiescently infected cells via the cyclic AMP signaling pathway. J Virol 2007; 81:6669-81. [PMID: 17301150 PMCID: PMC1900132 DOI: 10.1128/jvi.01524-06] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2006] [Accepted: 02/02/2007] [Indexed: 11/20/2022] Open
Abstract
The human cytomegalovirus (HCMV) major immediate-early (MIE) enhancer contains five functional cyclic AMP (cAMP) response elements (CRE). Because the CRE in their native context do not contribute appreciably to MIE enhancer/promoter activity in lytically infected human fibroblasts and NTera2 (NT2)-derived neurons, we postulated that they might have a role in MIE enhancer/promoter reactivation in quiescently infected cells. Here, we show that stimulation of the cAMP signaling pathway by treatment with forskolin (FSK), an adenylyl cyclase activator, greatly alleviates MIE enhancer/promoter silencing in quiescently infected NT2 neuronal precursors. The effect is immediate, independent of de novo protein synthesis, associated with the phosphorylation of ATF-1 serine 63 and CREB serine 133, dependent on protein kinase A (PKA) and the enhancer's CRE, and linked to viral-lytic-cycle advancement. Coupling of FSK treatment with the inhibition of either histone deacetylases or protein synthesis synergistically activates MIE gene expression in a manner suggesting that MIE enhancer/promoter silencing is optimally relieved by an interplay of multiple regulatory mechanisms. In contrast, MIE enhancer/promoter silence is not overcome by stimulation of the gamma interferon (IFN-gamma) signaling pathway, despite the enhancer having two IFN-gamma-activated-site-like elements. We conclude that stimulation of the cAMP/PKA signaling pathway drives CRE-dependent MIE enhancer/promoter activation in quiescently infected cells, thus exposing a potential mode of regulation in HCMV reactivation.
Collapse
Affiliation(s)
- Michael J Keller
- Department of Internal Medicine, University of Iowa Carver College of Medicine, and Veterans Affairs Medical Center, Iowa City, Iowa 52242, USA
| | | | | | | | | | | |
Collapse
|
14
|
Liew CG, Draper JS, Walsh J, Moore H, Andrews PW. Transient and stable transgene expression in human embryonic stem cells. Stem Cells 2007; 25:1521-8. [PMID: 17379764 DOI: 10.1634/stemcells.2006-0634] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Plasmid vectors remain a valuable yet capricious tool for the genetic manipulation of human embryonic stem (hES) cells. We have compared the efficacy of four promoters to mediate transient and stable transfection in hES and human embryonal carcinoma cell lines with the reporter enhanced green fluorescent protein (eGFP). In transient assays, the two mammalian promoters, UbiquitinC and Rosa26 (pUbiC and pR26), the human cytomegalovirus major immediate early promoter (HCMV-MIE; pCMV), and the HCMV-MIE/chicken beta-actin/rabbit beta-globin hybrid promoter (pCAGG) gave variable results that depended upon the cell line transfected but in an unpredictable way: each promoter supported strong transient expression in at least one cell line. The results for stable transfection were generally at variance with the transient assays. In each case, transgene silencing was quite marked, most notably with the pCMV, with which no eGFP-positive clones were obtained. An exception was the pCAG vector, in which the CAGG composite promoter is linked to the polyoma virus mutant enhancer PyF101; stable eGFP-positive transfectants were obtained, and these clones retained eGFP expression for over 120 passages, even in the absence of selection. However, if the PyF101 elements were removed, the resulting transfectants were also subjected to progressive gene silencing. Thus, the choice of promoter is critical for determining the desired effect of transgene expression in hES cells. Our data also demonstrate that pUbiC, pR26, pCAGG, and pCAG are more superior to the pCMV for generation of stable transfectants in hES cells. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Chee-Gee Liew
- Centre for Stem Cell Biology, Department of Biomedical Science, Western Bank, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | | | | | | | | |
Collapse
|
15
|
Abstract
Human cytomegalovirus (HCMV) persists as a subclinical, lifelong infection in the normal human host, maintained at least in part by its carriage in the absence of detectable infectious virus – the hallmark of latent infection. Reactivation from latency in immunocompromised individuals, in contrast, often results in serious disease. Latency and reactivation are defining characteristics of the herpesviruses and key to understanding their biology. However, the precise cellular sites in which HCMV is carried and the mechanisms regulating its latency and reactivation during natural infection remain poorly understood. This review will detail our current knowledge of where HCMV is carried in healthy individuals, which viral genes are expressed upon carriage of the virus and what effect this has on cellular gene expression. It will also address the accumulating evidence suggesting that reactivation of HCMV from latency appears to be linked intrinsically to the differentiation status of the myeloid cell, and how the cellular mechanisms that normally control host gene expression play a critical role in the differential regulation of viral gene expression during latency and reactivation.
Collapse
Affiliation(s)
- John Sinclair
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, UK
| | - Patrick Sissons
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, UK
| |
Collapse
|
16
|
Dósa R, Burián K, Gönczöl E. Human cytomegalovirus latency is associated with the state of differentiation of the host cells: an in vitro model in teratocarcinoma cells. Acta Microbiol Immunol Hung 2006; 52:397-406. [PMID: 16400879 DOI: 10.1556/amicr.52.2005.3-4.11] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The human cytomegalovirus (HCMV) major immediate-early (MIE) gene is not transcribed in undifferentiated NTera-2 embryonal carcinoma cells, but is transcribed in their differentiated derivatives, offering a model with which to study the developmental regulation of the activity of a viral gene during the differentiation of these cells. The molecular mechanisms involved in the blockade of the MIE gene expression in undifferentiated NTera2 cells include covalent closure of the circular conformation of the viral genome, silencing of the viral MIE promoter by histone deacetylation, and increases in the expression of negatively regulating transcription factors responsible for the recruitment of the histone deacytylases around the viral MIE promoter (MIEP), resulting in repression of the MIEP in undifferentiated cells. The treatment of NTera2 cells with retinoic acid induces the differentiation of these cells. In HCMV-infected differentiated NTera2 cells, the MIEP becomes associated with hyperacetylated histones, which results in an open structure of chromatin, enhancing the access of DNA-binding factors which positively regulate MIE gene expression and viral replication. This model system contributes to an understanding of HCMV latency and reactivation in vivo in the cells of the myeloid lineage.
Collapse
Affiliation(s)
- Réka Dósa
- Department of Dermatology, Sankt George County Hospital Fogolyán Kristóf, Sfintu Gheorghe, Romania
| | | | | |
Collapse
|
17
|
Ellsmore V, Reid GG, Stow ND. Detection of human cytomegalovirus DNA replication in non-permissive Vero and 293 cells. J Gen Virol 2003; 84:639-645. [PMID: 12604816 DOI: 10.1099/vir.0.18812-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human cytomegalovirus (HCMV) displays an exceptionally restricted host range in tissue culture with human fibroblasts being the principal fully permissive system. Nevertheless, immediate early (IE) proteins are expressed following infection of many non-permissive cell types of human, simian and murine origin, and viral origin-dependent DNA synthesis has been reconstituted by transfection of plasmids into Vero cells, a non-permissive line from African green monkey. We have examined the accumulation of HCMV strain AD169 DNA, and the replication of transfected HCMV origin-containing plasmids, in infected Vero and human embryonic kidney 293 cells, which were previously reported to express the major IE protein in a small proportion of infected cells but to be non-permissive for viral DNA synthesis. In Vero cells accumulation of origin-containing plasmid but not viral DNA occurred, whilst in 293 cells both DNAs accumulated. Immunofluorescence experiments indicated that following infection with 3 p.f.u. per cell, a small fraction of both cell types expressed the UL44 DNA replication protein. Neither cell line, however, supported the generation of infectious progeny virus. These results suggest that IE proteins expressed in Vero and 293 cells can induce the synthesis of early proteins capable of functioning in viral DNA replication, but there is a failure in later events on the pathway to infectious virus production. This provides further support for transfected Vero cells being a valid system in which to study HCMV DNA synthesis, and suggests that 293 cells may also prove useful in similar experiments.
Collapse
Affiliation(s)
- Victoria Ellsmore
- MRC Virology Unit, Institute of Virology, Church Street, Glasgow G11 5JR, UK
| | - G Gordon Reid
- MRC Virology Unit, Institute of Virology, Church Street, Glasgow G11 5JR, UK
| | - Nigel D Stow
- MRC Virology Unit, Institute of Virology, Church Street, Glasgow G11 5JR, UK
| |
Collapse
|
18
|
Abstract
BACKGROUND Reactivation of CMV from latency results in serious morbidity and mortality in immunocompromised transplant recipients. The mechanism by which CMV reactivates from latency has not been well understood. OBJECTIVE In this review we discuss three models for reactivation from latency and present evidence in favor of the model that reactivation is a multi-step process which is initiated by the allogeneic response to the transplanted organ. Study design (J. Virol. 75 (2001) 4814). Mice latently infected with murine cytomegalovirus (MCMV) were used as donors for allogeneic or syngeneic kidney transplants into immunocompetent recipients. The contralateral donor kidneys were used as controls. Transplanted kidneys were removed at various times after transplant and analyzed for expression of viral genes associated with productive infection and for expression of inflammatory cytokines. Electrophoretic mobility shift assay was performed on nuclear extracts of control and transplanted kidneys to examine activation of AP-1 and NFkappaB. Latently infected mice were also injected with tumor necrosis factor (TNF) to examine the effect of TNF alone on induction of MCMV immediate-early (IE) gene expression. Transgenic major immediate early promoter-lacZ mice carrying a beta-galactosidase reporter gene under the control of the human cytomegalovirus (HCMV) IE promoter/enhancer were used as donors for allogeneic kidney transplants to study the effect of allogeneic transplantation on induction of HCMV IE gene expression. RESULTS Allogeneic, but not syngeneic transplantation induces MCMV IE-1 expression and expression of inflammatory cytokines, including TNF. Allogeneic transplantation activates transcription factors, including NFkappaB and AP-1. TNF alone can induce MCMV IE-1 gene expression and activation of NFkappaB and AP-1 in some tissues. CONCLUSIONS We propose that induction of IE-1 gene expression is the first step in reactivation of the virus in an immunocompromised transplant recipient, and that it occurs as a result of the allogeneic response, which induces expression of TNF and subsequent activation of NFkappaB, and ischemia/reperfusion injury, which induces activation of AP-1. We speculate that the natural stimulus for reactivation in an immunocompetent host is an inflammatory immune response to infection and that allogeneic transplantation mimics this process.
Collapse
Affiliation(s)
- Mary Hummel
- Department of Surgery, Division of Organ Transplantation, Northwestern University Medical School, Chicago, IL 60611, USA.
| | | |
Collapse
|
19
|
Fortunato EA, Sanchez V, Yen JY, Spector DH. Infection of cells with human cytomegalovirus during S phase results in a blockade to immediate-early gene expression that can be overcome by inhibition of the proteasome. J Virol 2002; 76:5369-79. [PMID: 11991965 PMCID: PMC137046 DOI: 10.1128/jvi.76.11.5369-5379.2002] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cells infected with human cytomegalovirus (HCMV) after commencing DNA replication do not initiate viral immediate-early (IE) gene expression and divide before arresting. To determine the nature of this blockade, we examined cells that were infected 24 h after release from G(0) using immunofluorescence, laser scanning cytometry, and fluorescence-activated cell sorting (FACS) analysis. Approximately 40 to 50% of the cells had 2N DNA content, became IE(+) in the first 12 h, and arrested. Most but not all of the cells with >2N DNA content did not express IE antigens until after mitosis. To define the small population of IE(+) cells that gradually accumulated within the S and G(2)/M compartments, cells were pulsed with bromodeoxyuridine (BrdU) just prior to S-phase infection and analyzed at 12 h postinfection for IE gene expression, BrdU positivity, and cell cycle position. Most of the BrdU(+) cells were IE(-) and had progressed into G(2)/M or back to G(1). The majority of the IE(+) cells in S and G(2)/M were BrdU(-). Only a few cells were IE(+) BrdU(+), and they resided in G(2)/M. Multipoint BrdU pulse-labeling revealed that, compared to cells actively synthesizing DNA at the beginning of the infection, a greater percentage of the cells that initiated DNA replication 4 h later could express IE antigens and proceed into S. Synchronization of the cells with aphidicolin also indicated that the blockade to the activation of IE gene expression was established in cells soon after initiation of DNA replication. It appears that a short-lived protein in S-phase cells may be required for IE gene expression, as it is partially restored by treatment with the proteasome inhibitor MG132.
Collapse
Affiliation(s)
- Elizabeth A Fortunato
- Department of Microbiology, Molecular Biology and Biochemistry, University of Idaho, Moscow, Idaho 83844-3052, USA
| | | | | | | |
Collapse
|
20
|
Murphy JC, Fischle W, Verdin E, Sinclair JH. Control of cytomegalovirus lytic gene expression by histone acetylation. EMBO J 2002; 21:1112-20. [PMID: 11867539 PMCID: PMC125876 DOI: 10.1093/emboj/21.5.1112] [Citation(s) in RCA: 183] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Permissiveness for human cytomegalovirus (HCMV) infection is dependent on the state of cellular differentiation and has been linked to repression of the viral major immediate early promoter (MIEP). We have used conditionally permissive cells to analyze differential regulation of the MIEP and possible mechanisms involved in latency. Our data suggest that histone deacetylases (HDACs) are involved in repression of the MIEP in non-permissive cells as inhibition of HDACs induces viral permissiveness and increases MIEP activity. Non-permissive cells contain the class I HDAC, HDAC3; super-expression of HDAC3 in normally permissive cells reduces infection and MIEP activity. We further show that the MIEP associates with acetylated histones in permissive cells, and that in peripheral blood monocytes the MIEP associates with heterochromatin protein 1 (HP1), a chromosomal protein implicated in gene silencing. As monocytes are believed to be a site of viral latency in HCMV carriers and reactivated virus is only observed upon differentiation into macrophages, we propose that chromatin remodeling of the MIEP following cellular differentiation could potentially play a role in reactivation of latent HCMV.
Collapse
Affiliation(s)
- Jane C. Murphy
- Department of Medicine, University of Cambridge, Cambridge CB2 2QQ, UK and
Gladstone Institute of Virology and Immunology, University of California, San Francisco, CA, USA Corresponding author e-mail:
| | - Wolfgang Fischle
- Department of Medicine, University of Cambridge, Cambridge CB2 2QQ, UK and
Gladstone Institute of Virology and Immunology, University of California, San Francisco, CA, USA Corresponding author e-mail:
| | - Eric Verdin
- Department of Medicine, University of Cambridge, Cambridge CB2 2QQ, UK and
Gladstone Institute of Virology and Immunology, University of California, San Francisco, CA, USA Corresponding author e-mail:
| | | |
Collapse
|
21
|
Pizzorno MC. Nuclear cathepsin B-like protease cleaves transcription factor YY1 in differentiated cells. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1536:31-42. [PMID: 11335102 DOI: 10.1016/s0925-4439(01)00032-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Differentiation of pluripotent cells into differentiated cell types involves changes in many aspects of cellular biochemistry. Many of these changes result in alterations of gene expression, which may occur by changing the activity of transcription factors. The cell line NTERA-2 (NT2) can be differentiated into various cell types by incubation with retinoic acid. The differentiated cell type is also permissive for infection with the human herpesvirus cytomegalovirus (CMV). The transcription factor YY1 has been shown to regulate the immediate-early promoter of CMV in a differentiation specific manner by binding to one site at -958 to -950 and to at least two sites in the enhancer. It is demonstrated here that there is a second YY1 site in the modulator between -995 and -987. Levels of YY1 DNA binding activity and protein decrease in NT2 cells as they are differentiated with retinoic acid. This decrease in protein is due to the degradation of YY1 by a cathepsin B-like activity found in nuclear extracts. The cleavage products of YY1 include the intact C-terminal half of the protein, which contains the zinc fingers and the DNA binding activity. This suggests a mechanism that allows expression of the CMV immediate-early promoter in differentiated cells.
Collapse
Affiliation(s)
- M C Pizzorno
- Department of Biology, Bucknell University, Lewisburg, PA 17837, USA.
| |
Collapse
|
22
|
Meier JL. Reactivation of the human cytomegalovirus major immediate-early regulatory region and viral replication in embryonal NTera2 cells: role of trichostatin A, retinoic acid, and deletion of the 21-base-pair repeats and modulator. J Virol 2001; 75:1581-93. [PMID: 11160656 PMCID: PMC114067 DOI: 10.1128/jvi.75.4.1581-1593.2001] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2000] [Accepted: 11/14/2000] [Indexed: 01/29/2023] Open
Abstract
Inactivity of the human cytomegalovirus (HCMV) major immediate-early regulatory region (MIERR), which is composed of promoter, enhancer, unique region, and modulator, is linked to lack of HCMV replication in latently infected cells and in other nonpermissive cell types, including human embryonal NTera2 carcinoma (NT2) cells. I refined the embryonal NT2 cell model to enable characterization of the unknown mechanistic basis for silencing of HCMV MIERR-dependent transcription and viral replication in nonpermissive cells. These infected NT2 cells contain nonreplicating viral genomes with electrophoretic mobility equivalent to a supercoiled, bacterial artificial chromosome of comparable molecular weight. MIERR-dependent transcription is minimal to negligible. Increasing the availability of positive-acting transcription factors by retinoic acid (RA) treatment after infection is largely insufficient in reactivating the MIERR. In contrast, trichostatin A (TSA), a histone deacetylase inhibitor, reactivates MIERR-dependent transcription. Contrary to prior findings produced from transfected MIERR segments, deletion of the 21-bp repeats and modulator from the MIERR in the viral genome does not relieve MIERR silencing. To demonstrate that MIERR silencing likely results from enhancer inactivity, I examined an HCMV with a heterologous MIERR promoter that is enhancer dependent but exempt from IE2 p86-mediated negative autoregulation. This heterologous promoter, like its neighboring native MIERR promoter, exhibits immediate-early transcriptional kinetics in fibroblasts. In embryonal NT2 cells, the heterologous MIERR promoter is transcriptionally inactive. This silence is relieved by TSA but not by RA. Remarkably, TSA-induced reactivation of MIERR-dependent transcription from quiescent viral genomes is followed by release of infectious virus. I conclude that a mechanism of active repression imposes a block to MIERR-dependent transcription and viral replication in embryonal NT2 cells. Because TSA overcomes the block, viral gene silencing may involve histone deacetylase-based modification of viral chromatin, which might account for the covalently closed circular conformation of quiescent HCMV genomes.
Collapse
Affiliation(s)
- J L Meier
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, Iowa 52242, USA.
| |
Collapse
|
23
|
Bresnahan WA, Hultman GE, Shenk T. Replication of wild-type and mutant human cytomegalovirus in life-extended human diploid fibroblasts. J Virol 2000; 74:10816-8. [PMID: 11044129 PMCID: PMC110959 DOI: 10.1128/jvi.74.22.10816-10818.2000] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A cDNA encoding the catalytic subunit of human telomerase was used to generate life-extended derivatives of primary human diploid fibroblasts. The life-extended cells supported efficient human cytomegalovirus (HCMV) replication. A subclone of the life-extended cells was generated containing the HCMV UL82 gene and used to isolate and propagate a virus that exhibited a profound growth defect after infection at a low input multiplicity.
Collapse
Affiliation(s)
- W A Bresnahan
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544-1014, USA.
| | | | | |
Collapse
|
24
|
Sinclair J, Baillie J, Bryant L, Caswell R. Human cytomegalovirus mediates cell cycle progression through G(1) into early S phase in terminally differentiated cells. J Gen Virol 2000; 81:1553-65. [PMID: 10811939 DOI: 10.1099/0022-1317-81-6-1553] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Terminal differentiation of embryonal carcinoma cells and monocytes has been shown to be important for their permissiveness for human cytomegalovirus (HCMV) infection, even though such terminally differentiated cells have withdrawn from the cell cycle and are, essentially, in G(0) arrest. Recently, data from a number of laboratories have shown that productive infection with HCMV of quiescent fibroblasts held reversibly in G(0) of the cell cycle can result in cell cycle progression, which results eventually in cycle arrest. In contrast to quiescent fibroblasts, the effect of HCMV on cells that have withdrawn irreversibly from the cell cycle due to terminal differentiation has not, so far, been addressed. Here, it is shown that, in cells that have arrested in G(0) as a result of terminal differentiation, HCMV is able to induce cell functions associated with progression of the cell cycle through G(1) into early S phase. This progression is correlated with a direct physical and functional interaction between the HCMV 86 kDa major immediate-early protein (IE86) and the cyclin-dependent kinase inhibitor p21(Cip1).
Collapse
Affiliation(s)
- J Sinclair
- Department of Medicine, University of Cambridge, Level 5, Addenbrookes Hospital, Hills Road, Cambridge CB2 2QQ, UK.
| | | | | | | |
Collapse
|
25
|
Meier JL, Pruessner JA. The human cytomegalovirus major immediate-early distal enhancer region is required for efficient viral replication and immediate-early gene expression. J Virol 2000; 74:1602-13. [PMID: 10644329 PMCID: PMC111634 DOI: 10.1128/jvi.74.4.1602-1613.2000] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human cytomegalovirus (HCMV) major immediate-early (MIE) genes, encoding IE1 p72 and IE2 p86, are activated by a complex enhancer region (base positions -65 to -550) that operates in a cell type- and differentiation-dependent manner. The expression of MIE genes is required for HCMV replication. Previous studies analyzing functions of MIE promoter-enhancer segments suggest that the distal enhancer region variably modifies MIE promoter activity, depending on cell type, stimuli, or state of differentiation. To further understand the mechanism by which the MIE promoter is regulated, we constructed and analyzed several different recombinant HCMVs that lack the distal enhancer region (-300 to -582, -640, or -1108). In human fibroblasts, the HCMVs without the distal enhancer replicate normally at high multiplicity of infection (MOI) but replicate poorly at low MOI in comparison to wild-type virus (WT) or HCMVs that lack the neighboring upstream unique region and modulator (-582 or -640 to -1108). The growth aberrancy was normalized after restoring the distal enhancer in a virus lacking this region. For HCMVs without a distal enhancer, the impairment in replication at low MOI corresponds to a deficiency in production of MIE RNAs compared to WT or virus lacking the unique region and modulator. An underproduction of viral US3 RNA was also evident at low MOI. Whether lower production of IE1 p72 and IE2 p86 causes a reduction in expression of the immediate-early (IE) class US3 gene remains to be determined. We conclude that the MIE distal enhancer region possesses a mechanism for augmenting viral IE gene expression and genome replication at low MOI, but this regulatory function is unnecessary at high MOI.
Collapse
Affiliation(s)
- J L Meier
- Department of Internal Medicine and the Helen C. Levitt Center for Viral Pathogenesis and Disease, University of Iowa College of Medicine, Iowa City, Iowa 52242, USA.
| | | |
Collapse
|
26
|
Affiliation(s)
- E A Fortunato
- Department of Biology and Center for Molecular Genetics, University of California, San Diego, La Jolla 92093-0366, USA
| | | |
Collapse
|
27
|
Angulo A, Chandraratna RA, LeBlanc JF, Ghazal P. Ligand induction of retinoic acid receptors alters an acute infection by murine cytomegalovirus. J Virol 1998; 72:4589-600. [PMID: 9573222 PMCID: PMC109973 DOI: 10.1128/jvi.72.6.4589-4600.1998] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Here we report that administration of retinoids can alter the outcome of an acute murine cytomegalovirus (MCMV) infection. We show that a crucial viral control element, the major immediate-early enhancer, can be activated by retinoic acid (RA) via multiple RA-responsive elements (DR2) that bind retinoid X receptor-retinoic acid receptor (RAR) heterodimers with apparent dissociation constants ranging from 15 to 33 nM. Viral growth is dramatically increased upon RA treatment of infected tissue culture cells. Using synthetic retinoid receptor-specific agonists and antagonists, we provide evidence that RAR activation in cells is required for mediating the response of MCMV to RA. Oral administration of RA to infected immunocompetent mice selectively exacerbates an infection by MCMV, while cotreatment with an RAR antagonist protects against the adverse effects of RA on MCMV infection. In conclusion, these chemical genetic experiments provide evidence that an RAR-mediated pathway can modulate in vitro and in vivo infections by MCMV.
Collapse
Affiliation(s)
- A Angulo
- Departments of Immunology and Molecular Biology, Division of Virology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
28
|
Affiliation(s)
- P Ghazal
- Departments of Immunology and Neuropharmacology, Division of Virology R307B, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
29
|
Meier JL, Stinski MF. Effect of a modulator deletion on transcription of the human cytomegalovirus major immediate-early genes in infected undifferentiated and differentiated cells. J Virol 1997; 71:1246-55. [PMID: 8995648 PMCID: PMC191179 DOI: 10.1128/jvi.71.2.1246-1255.1997] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Differentiation-dependent expression of the human cytomegalovirus (HCMV) major immediate-early (MIE) genes, encoding IE1 and IE2, may partly govern virus replication in monocytic THP-1 and embryonal carcinoma (Tera-2) cells. The modulator of the MIE promoter was shown previously in transient transfection assays to repress transcription from promoter segments in undifferentiated THP-1 and Tera-2 cells but not in differentiated cells. To determine the biological importance of these findings, we constructed a recombinant HCMV (r delta MSVgpt) without a modulator. In comparison to wild-type (WT) virus, r delta MSVgpt exhibits a slight delay in growth in human fibroblasts, but there is no appreciable change in IE1 and IE2 transcription. Moreover, there is no appreciable change in the early/late kinetics of transcription of RNAs colinear with the predicted UL128 coding region, which is adjacent to the modulator, although the size distribution and abundance of these RNAs are altered. In infected undifferentiated THP-1 and Tera-2 cells, WT and r alpha MSVgpt viruses produce minimal but comparable amounts of IE1 RNAs. The genomes of both viruses are detectable in similar amounts within these undifferentiated cells. Induction of cellular differentiation before infection overcomes the block in MIE gene transcription. WT and r alpha MSVgpt infections of differentiated THP-1 cells produce similar levels of IE1 and IE2 RNAs. Thus, differentiation-dependent control of MIE gene transcription involves regulatory mechanisms other than the modulator. Possible alternative functions of the modulator are discussed.
Collapse
Affiliation(s)
- J L Meier
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City 52242, USA
| | | |
Collapse
|
30
|
Chan YJ, Chiou CJ, Huang Q, Hayward GS. Synergistic interactions between overlapping binding sites for the serum response factor and ELK-1 proteins mediate both basal enhancement and phorbol ester responsiveness of primate cytomegalovirus major immediate-early promoters in monocyte and T-lymphocyte cell types. J Virol 1996; 70:8590-605. [PMID: 8970984 PMCID: PMC190952 DOI: 10.1128/jvi.70.12.8590-8605.1996] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Cytomegalovirus (CMV) infection is nonpermissive or persistent in many lymphoid and myeloid cell types but can be activated in differentiated macrophages. We have shown elsewhere that both the major immediate-early gene (MIE) and lytic cycle infectious progeny virus expression can be induced in otherwise nonpermissive monocyte-like U-937 cell cultures infected with either human CMV (HCMV) or simian CMV (SCMV) by treatment with the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA). Two multicopy basal enhancer motifs within the SCMV MIE enhancer, namely, 11 copies of the 16-bp cyclic AMP response element (CRE) and 3 copies of novel 17-bp serum response factor (SRF) binding sites referred to as the SNE (SRF/NFkappaB-like element), as well as four classical NFkappaB sites within the HCMV version, contribute to TPA responsiveness in transient assays in monocyte and T-cell types. The SCMV SNE sites contain potential overlapping core recognition binding motifs for SRF, Rel/NFkappaB, ETS, and YY1 class transcription factors but fail to respond to either serum or tumor necrosis factor alpha. Therefore, to evaluate the mechanism of TPA responsiveness of the SNE motifs and of a related 16-bp SEE (SRF/ETS element) motif found in the HCMV and chimpanzee CMV MIE enhancers, we have examined the functional responses and protein binding properties of multimerized wild-type and mutant elements added upstream to the SCMV MIE or simian virus 40 minimal promoter regions in the U-937, K-562, HL-60, THP-1, and Jurkat cell lines. Unlike classical NFkappaB sites, neither the SNE nor the SEE motif responded to phosphatase inhibition by okadaic acid. However, the TPA responsiveness of both CMV elements proved to involve synergistic interactions between the core SRF binding site (CCATATATGG) and the adjacent inverted ETS binding motifs (TTCC), which correlated directly with formation of a bound tripartite complex containing both the cellular SRF and ELK-1 proteins. This protein complex was more abundant in U-937, K-562, and HeLa cell extracts than in Raji, HF, BALB/c 3T3, or HL-60 cells, but the binding activity was altered only twofold after TPA treatment. A 40-fold stimulation of chloramphenicol acetyltransferase activity mediated by four tandem repeats of the SNE could be induced within 2 h (and up to 250-fold within 6 h) after addition of TPA in DNA-transfected U-937 cells, indicating that the stimulation appeared likely to be a true protein kinase C-mediated signal transduction event rather than a differentiation response. Slight differences in the sequence of the core SRF binding site compared with that of the classical c-Fos promoter serum response element, together with differences in the spacing between the SRF and ETS motifs, appear to account for the inability of the SCMV SNEs to respond to serum induction.
Collapse
Affiliation(s)
- Y J Chan
- Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | |
Collapse
|
31
|
Chan YJ, Tseng WP, Hayward GS. Two distinct upstream regulatory domains containing multicopy cellular transcription factor binding sites provide basal repression and inducible enhancer characteristics to the immediate-early IES (US3) promoter from human cytomegalovirus. J Virol 1996; 70:5312-28. [PMID: 8764042 PMCID: PMC190489 DOI: 10.1128/jvi.70.8.5312-5328.1996] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The US3 gene of human cytomegalovirus (HCMV) is expressed at immediate-early (IE) times in permissive HF cells, but not in nonpermissive rodent cells, and encodes several proteins that have been reported to have regulatory characteristics, although they are dispensable for growth in cell culture. Both spliced and unspliced forms of US3 IE transcripts are associated with the second of only two known large and complex upstream enhancer domains within the 229-kb HCMV genome, which we refer to as the IES cis-acting control region. Only the 260-bp proximal segment (from -313 to -55) of the 600-bp IES control domain, which contains multicopy NF-kappaB binding sites, proved to be necessary to transfer both high basal expression plus phorbol ester- and okadaic acid-inducible characteristics to heterologous promoters in transient assays in U-937 and K-562 cells. However, the IES control region contains a distinctive 280-bp distal domain, characterized by the presence of seven interspersed repeats of a 10-bp TGTCGCGACA palindromic consensus motif that encompasses a NruI site. This far upstream Nru repeat region (from -596 to -314) imparted up to 20-fold down-regulation effects onto strong basal heterologous promoters as well as onto the IES enhancer plus minimal promoter region in both U-937 and K-562 cells. Functional Nru repressor elements (NREs) could not be generated by multimerizing either the palindromic (P) Nru motifs alone or adjacent degenerate interrupted (I Nru motifs alone. However, multimerized forms of the combined P plus I elements reconstituted the full 20-fold cis-acting down-regulation phenotype of the intact NRE domain. The P and I forms of the Nru elements each bound independently and specifically to related cellular DNA-binding factors to form differently migrating A or B complexes, respectively, whereas the combined P plus I elements bound cooperatively to both the A and B complexes with high affinity. Interestingly, nuclear extracts from U-937, K-562, HeLa, and Vero cells all formed both the A and B NRE binding factor complexes, whereas those from HF cells produced only A complexes, and Raji, HL60, and BALB/c 3T3 cells lacked both types of binding factor complexes. The core pentameric CGACA and CGATA half sites present in both the P and I Nru motifs are related to recently described Drosophila chromosomal insulator binding sites. Therefore, in addition to its cis-repression or silencer characteristics, the NRE domain appears likely to act to shield adjacent segments of the viral genome from the chromatin-reorganizing effects of the IES-inducible enhancer. We speculate that differential expression and regulation of the IES enhancer-controlled US3 protein, either in concert with or separately from the major IE (MIE) enhancer-controlled IE1 and IE2 transactivator proteins, may play a critical role in determining HCMV permissiveness in some cell types and perhaps also in the establishment of or reactivation from latency.
Collapse
Affiliation(s)
- Y J Chan
- The Molecular Virology Laboratories, Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | |
Collapse
|
32
|
Huang TH, Oka T, Asai T, Okada T, Merrills BW, Gertson PN, Whitson RH, Itakura K. Repression by a differentiation-specific factor of the human cytomegalovirus enhancer. Nucleic Acids Res 1996; 24:1695-701. [PMID: 8649988 PMCID: PMC145859 DOI: 10.1093/nar/24.9.1695] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
We detected a novel nuclear protein, MRF, that binds to multiple sites on the modulator which is located upstream of the human cytomegalovirus major immediate early gene enhancer. The expression of MRF is differentiation specific; the DNA binding activity is present in nuclear extracts from undifferentiated Tera-2 and THP-1 cells, but significantly reduced after these cells are induced to differentiate. In undifferentiated cells the enhancer activity is repressed by the modulator and upon differentiation the enhancer becomes active. Competitive binding assays demonstrate that MRF requires the presence of multiple A+T stretches for binding to DNA, rather than binding to a specific DNA sequence. Mutations of these stretches in the modulator reduce the binding activity of MRF, as well as the repressing activity on the enhancer. These results suggest that MRF may act as a repressor of enhancer function. We propose that MRF binds over the entire modulator and exerts repressor activity.
Collapse
Affiliation(s)
- T H Huang
- Department of Molecular Genetics, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Plachter B, Sinzger C, Jahn G. Cell types involved in replication and distribution of human cytomegalovirus. Adv Virus Res 1996; 46:195-261. [PMID: 8824701 DOI: 10.1016/s0065-3527(08)60073-1] [Citation(s) in RCA: 129] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
As the number of patients suffering from severe HCMV infections has steadily increased, there is a growing need to understand the molecular mechanisms by which the virus causes disease. The factors that control infection at one time and the events leading to virus multiplication at another time are only beginning to be understood. The interaction of HCMV with different host cells is one key for elucidating these processes. Through modern techniques, much has been learned about the biology of HCMV infections in culture systems. In addition to endothelial cells, epithelial cells, and smooth muscle cells, fibroblasts are one cell population preferentially infected in solid tissues in vivo. From these sites of multiplication, the virus may be carried by peripheral monocytes and circulating endothelial cells to reach distant sites of the body. This would explain the multiorgan involvement in acute HCMV infection and the modes of viral transmission. From what has been learned mainly from human fibroblast culture systems, future studies will focus on how HCMV regulates the expression of its putative 200 genes in different host cells at different stages of cell differentiation and activation to result in viral latency and pathogenesis.
Collapse
Affiliation(s)
- B Plachter
- Institut für Klinische und Molekulare Virologie, Universität Erlangen-Nürnberg, Germany
| | | | | |
Collapse
|
34
|
Ramesh N, Shin YK, Lau S, Osborne WR. High-level expression from a cytomegalovirus promoter in macrophage cells. Hum Gene Ther 1995; 6:1323-7. [PMID: 8590737 DOI: 10.1089/hum.1995.6.10-1323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
To identify vectors that provide optimal gene expression in human hematopoietic cells, we investigated retroviruses containing the adenosine deaminase (ADA) gene under the transcriptional control of the promoters/enhancers of Moloney murine leukemia virus and the human cytomegalovirus (CMV) immediate-early gene. ADA expression was monitored in transduced human multipotential promyelocytic leukemic HL-60 cells and human monocytic leukemic THP-1 cells. HL-60 cells can be induced by phorbol ester to differentiate into macrophage lineage cells and by retinoic acid into granulocytic cells. THP-1 cells undergo phorbol ester-induced differentiation to macrophage cells. In LNCA-transduced HL-60 derived macrophage cells, ADA controlled by the CMV promoter was expressed at 100.0 mumol/hr.mg, in contrast to 1.2 mumol/hr.mg from LN-transduced control cells. LNCA-transduced THP-1 macrophage cells showed a similar increase in ADA activity over control cells. These elevated enzyme activities were confirmed by Northern blots, which showed substantial increases in ADA mRNA derived from the CMV promoter. This suggests use of the CMV promoter for gene therapy targeted at macrophages, as, for example, in the treatment of lysosomal storage disorders such as Gaucher disease. These inducible cell lines have allowed the evaluation of transduced gene expression in proliferating and differentiating hematopoietic cells that may serve as an in vitro model of bone marrow-targeted gene therapy.
Collapse
Affiliation(s)
- N Ramesh
- Department of Pediatrics, University of Washington, Seattle 98195, USA
| | | | | | | |
Collapse
|
35
|
Chang YN, Jeang KT, Lietman T, Hayward G. Structural Organization of the Spliced Immediate-Early Gene Complex that Encodes the Major Acidic Nuclear (IE1) and Transactivator (IE2) Proteins of African Green Monkey Cytomegalovirus. J Biomed Sci 1995; 2:105-130. [PMID: 11725047 DOI: 10.1007/bf02253062] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Total immediate-early (IE) RNA synthesized after infection with African green monkey cytomegalovirus (SCMV) in the presence of cycloheximide contained a major 2.3-kb mRNA species that acted as template for in vitro synthesis of a single 94-kD nuclear protein. The same IE RNA hybridized predominantly to a 1.8-kb subregion of the 220-kb genome which mapped 1.5 kb to the left of the in vitro transcription start site and TATATAA motif previously associated with the powerful MIE (IE94) enhancer region. However, DNA sequence and S1-mapping analysis of a 5-kb region downstream from the promoter revealed the existence of a far upstream noncoding first exon and four additional spliced exons capable of encoding two alternative protein products with shared N-terminal domains. This region is similar in structure to that of the MIE gene complex of human cytomegalovirus (HCMV), including being highly CpG suppressed. Exons 2, 3, and 4 encode an acidic protein equivalent to the 68-kD IE1 protein (UL123) of HCMV and exons 2, 3, and 5 encode a protein equivalent to the 80-kD IE2 (UL122) DNA-binding protein of HCMV. Transcripts from across the IE2 region were detected within the cycloheximide RNA, but they were present at 10- to 20-fold lower abundance than IE1 transcripts. The proposed 547-codon IE1 (IE94) acidic phosphoprotein of SCMV displays minimal residual homology with the IE1 protein of HCMV, but both associate with metaphase chromosomes and have large C-terminal glutamic-acid-rich domains. In contrast, the proposed 583-codon IE2 protein of SCMV displays extensive amino acid similarity to the HCMV IE2 transcriptional regulatory protein especially within C-terminal domains that are known to play a major role in promoter targeting for both transactivation and negative autoregulation functions. Copyright 1995 S. Karger AG, Basel
Collapse
Affiliation(s)
- Y.-N. Chang
- The Molecular Virology Laboratories, Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Md., USA
| | | | | | | |
Collapse
|
36
|
Furukawa T, Jisaki F, Sakamuro D, Takegami T, Murayama T. Detection of human cytomegalovirus genome in uterus tissue. Arch Virol 1994; 135:265-77. [PMID: 7979967 DOI: 10.1007/bf01310013] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
To analyze persistent infection by human cytomegalovirus (HCMV) in vivo, specimens obtained from various sources and autopsied organs were examined for the presence of HCMV DNA, mRNA transcripts and antigens by polymerase chain reaction, in situ hybridization and immunostaining. The HCMV genome was detected in lung, liver, kidney, and blood vessels at an average positive rate of 15%. The highest PCR-positive rate was observed with cervical smears. Subsequent examination of uterus tissues from patients with myoma revealed HCMV transcripts and antigens in glandular epithelial cells, leukocytes, endothelial cells, and others, indicating productive HCMV infection of cervical tissue.
Collapse
Affiliation(s)
- T Furukawa
- Department of Microbiology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | | | | | | | | |
Collapse
|
37
|
Liu R, Baillie J, Sissons JG, Sinclair JH. The transcription factor YY1 binds to negative regulatory elements in the human cytomegalovirus major immediate early enhancer/promoter and mediates repression in non-permissive cells. Nucleic Acids Res 1994; 22:2453-9. [PMID: 8041605 PMCID: PMC308195 DOI: 10.1093/nar/22.13.2453] [Citation(s) in RCA: 106] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
We have previously shown that repression of human cytomegalovirus (HCMV) major immediate early (IE) gene expression in non-permissive human teratocarcinoma (T2) cells is associated with a number of nuclear factors which bind to the imperfect dyad symmetry located in the modulator region upstream of the major IE enhancer as well as to the 21 bp repeat elements within the enhancer. Differentiation of T2 cells with retinoic acid (RA) results in a decrease in binding of some of these nuclear factors to these sites and deletion of these specific binding sites from major IE promoter/reporter constructs results in increased IE promoter activity in normally non-permissive cells. In this study, we demonstrate that the transcription factor YY1, which can negatively regulate the adeno-associated virus P5 promoter, directly binds to both the imperfect dyad symmetry and the 21 bp repeat elements in the HCMV major IE promoter/regulatory region and mediates repression of HCMV IE gene expression. This strongly suggests that YY1 plays an important role in regulating HCMV expression in non-permissive cells.
Collapse
Affiliation(s)
- R Liu
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, UK
| | | | | | | |
Collapse
|
38
|
Rideg K, Hirka G, Prakash K, Bushar LM, Nothias JY, Weinmann R, Andrews PW, Gönczöl E. DNA-binding proteins that interact with the 19-base pair (CRE-like) element from the HCMV major immediate early promoter in differentiating human embryonal carcinoma cells. Differentiation 1994; 56:119-29. [PMID: 8026641 DOI: 10.1046/j.1432-0436.1994.56120119.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The pluripotent human embryonal carcinoma (EC) cell line NTERA-2 provides a useful tool for investigating cell differentiation in a way that is pertinent to the development of the early human embryo. The major immediate early (MIE) gene of human cytomegalovirus (HCMV), which is not transcribed in undifferentiated NTERA-2 EC cells but is transcribed in their differentiated derivatives, offers a model with which to study the developmental regulation of gene activity during the differentiation of these cells. We have investigated the regulatory activity of the cAMP response elements (CRE) and the activation protein (AP1) site found within several repeated 19-base-pair (bp) elements from the HCMV MIE promoter, and the developmental regulation of nuclear DNA-binding factors that interact with these sites. The 19-bp CRE but not the AP1 site is responsive to cAMP in undifferentiated NTERA-2 EC and its activity is enhanced upon differentiation. Nuclear proteins of the CREB, Fos, and Jun families bind to these sites, but, surprisingly, their levels only show limited regulation during NTERA-2 differentiation. This contrasts with results obtained with murine EC cells. However, additional and apparently novel proteins with molecular weights between 80,000 and 90,000, and binding specificities for both CRE and AP1 sites, were detected in undifferentiated EC cells. The activity of these proteins decreased markedly after differentiation, indicating their involvement in negative regulation of the CRE/AP1-like site in undifferentiated EC cells. This suggests novel members able to interact via leucine zippers with other members of the Jun-Fos-CREB family of DNA binding proteins that are also involved in this regulation.
Collapse
Affiliation(s)
- K Rideg
- Wistar Institute of Anatomy and Biology, Philadelphia, PA 19104
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Chiou CJ, Zong J, Waheed I, Hayward GS. Identification and mapping of dimerization and DNA-binding domains in the C terminus of the IE2 regulatory protein of human cytomegalovirus. J Virol 1993; 67:6201-14. [PMID: 8396676 PMCID: PMC238042 DOI: 10.1128/jvi.67.10.6201-6214.1993] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The 80-kDa IE2 nuclear phosphoprotein encoded by the human cytomegalovirus (HCMV) major immediate-early (MIE) gene behaves both as a nonspecific transactivator of heterologous reporter genes and as a specific repressor of its own promoter-enhancer region. To begin to examine the biochemical properties of the IE2 protein, we prepared panels of N-terminal and C-terminal truncation mutants by in vitro translation procedures. In cross-linking experiments, the C-terminal half of IE2 (which is sufficient for down-regulation) formed dimers but N-terminal segments did not do so. Cotranslated Oct2/IE2 fusion proteins containing the same IE2 C-terminal region from codons 266 to 579 also formed mixed-subunit DNA-bound oligomeric complexes in gel mobility shift assays. Furthermore, an IE2 domain bounded by codons 388 to 542 proved to immunoprecipitate as heterodimers with cotranslated subunits containing known epitopes for specific antibodies. Deletion up to codon 428 or truncation back to codon 504 prevented this interaction. In direct gel shift DNA-binding assays, a bacterial GST/IE2(346-579) fusion protein bound to a 30-mer oligonucleotide probe encompassing the major immediate-early gene negative cis-regulatory target DNA sequence but failed to bind to a single-base-pair insertion mutant probe (delta CRS). This specific DNA-binding activity was abolished by further deletion up to codon 388 on the N-terminal side or by truncation at codon 542 on the C-terminal side. Therefore, the minimal DNA-binding domain requires additional amino acid motifs on both sides of the dimerization domain. This segment of IE2 is functionally important for both transactivation and down-regulation and contains several highly conserved amino acid motifs that are shared amongst the equivalent HCMV, simian CMV, mouse CMV, rat CMV, and human herpesvirus 6 proteins from other betaherpesviruses.
Collapse
Affiliation(s)
- C J Chiou
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205-2185
| | | | | | | |
Collapse
|
40
|
Liang GS, Daikos GL, Serfling U, Zhu WY, Pecoraro M, Leonardi CL, Fischl MA, Penneys NS. An evaluation of oral ulcers in patients with AIDS and AIDS-related complex. J Am Acad Dermatol 1993; 29:563-8. [PMID: 8408791 DOI: 10.1016/0190-9622(93)70222-f] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Patients with HIV infection can have recurrent and persistent oral ulcers, not attributable to known infectious agents. OBJECTIVE Our aim was to evaluate prospectively oral ulcers in patients with HIV infection to determine whether an etiologic agent could be identified. METHODS Sixteen patients with HIV infection who had oral ulcers not attributable to known causes had culture of the base and a biopsy specimen taken from the ulcer. Cultures were obtained for herpes simplex and varicella-zoster viruses, mycobacteria, and fungi. By polymerase chain reaction (PCR) analysis with primer/probe sets for herpes simplex viruses 1 and 2, varicella-zoster virus, cytomegalovirus, human papillomavirus, and Mycobacterium tuberculosis, each biopsy specimen was analyzed for the presence of DNA from these organisms. Specimens were also evaluated histologically. RESULTS Histoplasmosis was detected histologically in one biopsy specimen, candidiasis in a second, and herpetic changes in a third. Viral cultures were positive for herpes simplex virus 1 in four cases and herpes simplex virus 2 in one case. PCR analysis detected DNA for herpes simplex virus 1 in one case and herpes simplex virus 2 in another; DNA from other pathogens was not identified. In the remaining eight patients, hematoxylin-and-eosin staining revealed eosinophilic ulcers in five cases and nonspecific changes in three cases. CONCLUSION The etiologic agent of recurrent or persistent oral ulcers in patients with AIDS and AIDS-related complex was not identified in 50% of patients. PCR analysis was not useful. Herpes simplex virus or other pathogens were not detected in ulcers containing numerous eosinophils.
Collapse
Affiliation(s)
- G S Liang
- Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine, Florida
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Human foreskin fibroblasts (HFF) were immortalized via retrovirus-mediated gene transfer of the E6 and E7 genes of human papillomavirus type 16. An immortalized fibroblast (IF) cell line which was morphologically akin to the parental cell line was isolated. The IF cell line was evaluated for permissiveness to human cytomegalovirus (HCMV) infection after the IF cell line surpassed the normal passage limitation of diploid fibroblasts. Western immunoblot analysis of representative HCMV-encoded immediate-early (72-kDa), early (gB), and late (gH) gene products demonstrated that the IF cell line produced these proteins analogous to those produced by the parental HFF cells. Similar quantities of infectious virus were produced in the IF and HFF cell lines as determined in one-step growth curve experiments. Compared with the HFF cells, morphologically identical plaques were produced in the IF cell line in approximately 10 to 12 days postinfection. These findings indicate that fibroblast cell lines immortalized with transforming genes of human papillomavirus retain complete permissiveness to HCMV infection and support plaque formation. The IF cell line will be useful for future genetic analysis of HCMV.
Collapse
Affiliation(s)
- T Compton
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison Medical School 53706-1532
| |
Collapse
|
42
|
Chang YN, Jeang KT, Chiou CJ, Chan YJ, Pizzorno M, Hayward GS. Identification of a large bent DNA domain and binding sites for serum response factor adjacent to the NFI repeat cluster and enhancer region in the major IE94 promoter from simian cytomegalovirus. J Virol 1993; 67:516-29. [PMID: 8380090 PMCID: PMC237389 DOI: 10.1128/jvi.67.1.516-529.1993] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The major immediate-early (MIE) transactivator proteins of cytomegaloviruses (CMV) play a pivotal role in the initiation of virus-host cell interactions. Therefore, cis- and trans-acting factors influencing the expression of these proteins through their upstream promoter-enhancer regions are important determinants of the outcome of virus infection. S1 nuclease analysis and in vitro transcription assays with the MIE (or IE94) transcription unit of simian CMV (SCMV) (Colburn) revealed a single prominent mRNA start site associated with a canonical TATATAA motif. This initiator region lies adjacent to a 2,400-bp 5'-upstream noncoding sequence that encompasses a newly identified 1,000-bp (A+T)-rich segment containing intrinsically bent DNA (domain C), together with the previously described proximal cyclic AMP response element locus (domain A) and a tandemly repeated nuclear factor I binding site cluster (domain B). Deleted MIE reporter gene constructions containing domain A sequences only yield up to 4-fold stronger basal expression in Vero cells than the intact simian virus 40 promoter-enhancer region, and sequences from position -405 to -69 (ENH-A1) added to a minimal heterologous promoter produced a 50-fold increase of basal expression in an enhancer assay. In contrast, neither the nuclear factor I cluster nor the bent DNA region possessed basal enhancer properties and neither significantly modulated the basal activity of the ENH-A1 segment. A second segment of domain A from position -580 to -450 was also found to possess basal enhancer activity in various cell types. This ENH-A2 region contains three copies of a repeated element that includes the 10-bp palindromic sequence CCATATATGG, which resembles the core motif of serum response elements and proved to bind specifically to the cellular nuclear protein serum response transcription factor. Reporter gene constructions containing four tandem copies of these elements displayed up to 13-fold increased basal enhancer activity and 18-fold tetradecanoyl phorbol acetate responsiveness in U937 cells, but an ENH-A2 DNA segment encompassing two of the core serum response transcription factor binding sites failed to respond to serum induction in NIH 3T3 cells. Although there are overall similarities in the organizations of both the MIE enhancers and MIE transcription units among human CMV, SCMV, and murine CMV, the specific arrangements of repetitive motifs are quite different, and the bent DNA and ENH-A2 domains appear to be unique to SCMV.
Collapse
Affiliation(s)
- Y N Chang
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | | | | | | | | | | |
Collapse
|
43
|
Transcription Factors and Viral Regulatory Proteins as Potential Mediators of Human Cytomegalovirus Pathogenesis. MOLECULAR ASPECTS OF HUMAN CYTOMEGALOVIRUS DISEASES 1993. [DOI: 10.1007/978-3-642-84850-6_18] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
44
|
Mazeron MC, Benjelloun B, Bertrand C, Pons JL, Pérol Y. Comparison of MRC-5 and continuous cell lines for detection of cytomegalovirus in centrifugation cultures. J Virol Methods 1992; 39:311-7. [PMID: 1331154 DOI: 10.1016/0166-0934(92)90104-l] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Continuous cell lines were assessed for use for rapid human cytomegalovirus (HCMV) detection procedures combining tissue culture, centrifugation, and immediate early antigen (IEA) immunostaining. Human cells (MRC-5 embryonic fibroblasts, U-373MG astrocytoma cells, differentiated teratocarcinoma (Tera-2) cells), murine cells (BALB/c-3T3 and Y-1 cells), BHK21 hamster cells, and mink lung (ML) cells were first inoculated with HCMV laboratory strain. IEA synthesizing cells were detected by immunoperoxidase assay using a monoclonal antibody. ML cells and differentiated Tera-2 cells exhibited more positive cells than MRC-5 cells. BHK21, and MRC-5 cells were equivalent in sensitivity whereas U-373MG, BALB/c-3T3, and Y-1 cells had only reduced IEA positive cells. When 63 urine specimens were inoculated onto MRC-5, ML and differentiated Tera-2 cells, 20 (31.7%) were positive in MRC-5 cells versus 18 (28.5%) in ML or Tera-2 cells. Moreover, greater numbers of infected cells were detected in MRC-5 cells than in these two cell lines. MRC-5 cells were superior for detection of HCMV in clinical samples by centrifugation cultures.
Collapse
Affiliation(s)
- M C Mazeron
- Service de Bactériologie-Virologie, Hôpital Saint-Louis, Université Paris VII, France
| | | | | | | | | |
Collapse
|
45
|
Ghazal P, DeMattei C, Giulietti E, Kliewer SA, Umesono K, Evans RM. Retinoic acid receptors initiate induction of the cytomegalovirus enhancer in embryonal cells. Proc Natl Acad Sci U S A 1992; 89:7630-4. [PMID: 1323848 PMCID: PMC49764 DOI: 10.1073/pnas.89.16.7630] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Reactivation of latent virus is believed to result from a signal transduction event that induces immediate-early (IE) gene transcription. Evidence is presented that the major IE promoter (MIEP) of human cytomegalovirus (hCMV) is activated by physiological levels of retinoic acid (RA) in human embryonal carcinoma cells. Mutagenesis experiments localized in the MIEP enhancer, a retinoic acid-responsive element composed of a direct repeat separated by five nucleotides. Protein-DNA binding experiments revealed that this element functions as a specific target site for the direct interaction of nuclear receptor proteins for RA. These findings implicate the biologically active derivative of vitamin A (RA) as a potential modulator of hCMV pathogenesis in infants and immunocompromised adults.
Collapse
Affiliation(s)
- P Ghazal
- Department of Immunology, Scripps Research Institute, La Jolla, CA 92037
| | | | | | | | | | | |
Collapse
|
46
|
Tanaka J, Sadanari H, Sato H, Fukuda S. Sodium butyrate-inducible replication of human cytomegalovirus in a human epithelial cell line. Virology 1991; 185:271-80. [PMID: 1656587 DOI: 10.1016/0042-6822(91)90774-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Replication of human cytomegalovirus (HCMV) in a human epithelial thyroid papillary carcinoma cell line (TPC-1) was restricted. However, pretreatment of these cells with 5 mM sodium butyrate (NaB) for 24 hr before infection enhanced both HCMV yield and infectious center titer to a similar level of that seen in human embryonic lung fibroblast cells. Immunofluorescence staining, gel electrophoresis, and Northern blot analysis revealed that TPC-1 cells are nonpermissive for expression of HCMV major immediate early (IE1) functions, but many of the cells become permissive after being treated with NaB. The presence of cycloheximide during NaB pretreatment of the cells efficiently diminished the stimulatory effect of NaB on expression of the IE1 gene. Therefore, it appeared that NaB induces the synthesis of a cellular protein(s) which apparently plays an important role in the conversion of nonpermissive cells to a permissive state for expression of this critical viral gene. Transient chloramphenicol acetyltransferase (CAT) assay experiments indicated that in TPC-1 cells the HCMV-CAT construct which contains the complete IE1 promoter regulatory region was expressed poorly, whereas a high level of CAT activity was detectable in the NaB-treated cells. Therefore, these results suggest that the enhancing effect of NaB on HCMV replication is expressed through some host cellular factor(s), and the HCMV IE1 promoter regulatory region is most likely to be the primary target of NaB action.
Collapse
MESH Headings
- Antibodies, Monoclonal
- Antigens, Viral/analysis
- Butyrates/pharmacology
- Butyric Acid
- Carcinoma, Papillary
- Cell Line
- Cytomegalovirus/drug effects
- Cytomegalovirus/genetics
- Cytomegalovirus/physiology
- Dose-Response Relationship, Drug
- Electrophoresis, Polyacrylamide Gel
- Epithelium
- Fluorescent Antibody Technique
- Humans
- Lung
- Plasmids
- Promoter Regions, Genetic
- Protein Biosynthesis/drug effects
- RNA, Viral/biosynthesis
- RNA, Viral/genetics
- Regulatory Sequences, Nucleic Acid
- Thyroid Neoplasms
- Transcription, Genetic/drug effects
- Viral Proteins/analysis
- Viral Proteins/biosynthesis
- Virus Replication/drug effects
Collapse
Affiliation(s)
- J Tanaka
- Department of Virology, Kanazawa University, Japan
| | | | | | | |
Collapse
|
47
|
Kothari S, Baillie J, Sissons JG, Sinclair JH. The 21bp repeat element of the human cytomegalovirus major immediate early enhancer is a negative regulator of gene expression in undifferentiated cells. Nucleic Acids Res 1991; 19:1767-71. [PMID: 1851560 PMCID: PMC328102 DOI: 10.1093/nar/19.8.1767] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The major immediate early regulatory region of human cytomegalovirus (HCMV) has a complex set of DNA sites through which both cellular and viral factors coordinately regulate immediate early gene expression. In undifferentiated human teratocarcinoma (T2) cells we have previously shown that major immediate early gene expression is repressed by a differentiation specific nuclear factor MBF1, which binds to the imperfect dyad symmetry located upstream of the enhancer. However, upon differentiation MBF1 decreases resulting in immediate early gene expression. In this study we show, by mobility shift analysis that the same or similar factor(s) also binds to the 21bp repeat of the major immediate early enhancer. Deletion of this 21bp repeat from the immediate early enhancer expression vectors results in increased CAT expression in undifferentiated T2 cells, to levels similar to that in differentiated cells. Consequently, the 21bp repeat of the HCMV enhancer also acts to negatively regulate major immediate early enhancer function in non-permissive cells.
Collapse
Affiliation(s)
- S Kothari
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, UK
| | | | | | | |
Collapse
|
48
|
Andrews PW, Marrink J, Hirka G, von Keitz A, Sleijfer DT, Gönczöl E. The surface antigen phenotype of human embryonal carcinoma cells: modulation upon differentiation and viral infection. Recent Results Cancer Res 1991; 123:63-83. [PMID: 1660624 DOI: 10.1007/978-3-642-84485-0_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- P W Andrews
- Wistar Institute of Anatomy and Biology, Philadelphia, PA 19104
| | | | | | | | | | | |
Collapse
|
49
|
Pizzorno MC, Hayward GS. The IE2 gene products of human cytomegalovirus specifically down-regulate expression from the major immediate-early promoter through a target sequence located near the cap site. J Virol 1990; 64:6154-65. [PMID: 2173785 PMCID: PMC248790 DOI: 10.1128/jvi.64.12.6154-6165.1990] [Citation(s) in RCA: 139] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The 82-kDa IE2 protein of human cytomegalovirus (HCMV) acts as both a powerful nonspecific trans activator of heterologous promoters and a negative autoregulator of HCMV immediate-early gene expression in transient assays. We show here that the highly specific down-regulation effect occurs in permissive diploid human fibroblast cells as well as in nonpermissive Vero cells and that the target sequences are conserved within the major immediate-early promoters of both HCMV and simian cytomegalovirus. The response sequences were localized between -67 and +30 in the simian cytomegalovirus IE94 promoter and upstream of position +9 in the HCMV IE68 promoter. Deletion of sequences downstream of -14 in a target IE68-CAT gene abolished the negative phenotype and resulted in a reporter gene that was stimulated instead of inhibited by cotransfection with IE2 effector DNA. Insertion of an oligonucleotide containing sequences from between -17 and +9 into the IE68-CAT deletion construction restored autoregulation in either orientation. Furthermore, this same oligonucleotide transferred the full down-regulation phenotype when inserted at +10 into the nonresponsive IE175 promoter from herpes simplex virus. Therefore, a specific response signal that acts at the DNA level must lie within these boundaries. Additional analysis with inserted oligonucleotides containing deletions or point mutations revealed that essential components of the signal lie between positions -12 and +5. Therefore, negative autoregulation by HCMV IE2 in DNA cotransfection systems resembles that for simian virus 40 large T antigen and herpes simplex virus IE175 by acting through a signal located near the cap site, but the target sequence itself bears no resemblance to those utilized in these other viral systems.
Collapse
Affiliation(s)
- M C Pizzorno
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | | |
Collapse
|
50
|
Affiliation(s)
- J D Benson
- Curriculum in Genetics, University of North Carolina, Chapel Hill
| | | |
Collapse
|