1
|
Böttiger D, Öberg B. Effect of Herpesvirus Inhibition on Primary SIV Infection in Cynomolgus Monkeys. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/095632029600700306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Foscarnet and (-)9-[4-hydroxy-2-(hydroxymethyl)butyl] guanine (H2G) have already been shown to inhibit herpesviruses in vitro and also to inhibit viral antigen production in primary SIV infection in monkeys. Attempts have been made to determine if these invivo effects on SIV were due to a direct effect on SIV or were mediated through inhibition of endogenous transactivating herpesviruses. The possible involvement of herpesviruses in primary SIVsm infection in monkeys was studied by the use of various inhibitors of herpesvirus replication. Subcutaneous injections of 3 × 5 mg kg−1 day−1 of aciclovir, 3 × 5 mg kg−1 day−1 of ganciclovir and 3 × 28 mg kg−1 day−1 of phosphonoacetic acid had no effect on primary SIVsm infection in cynomolgus monkeys. These doses of aciclovir, ganciclovir and phosphonoacetic acid are inhibitory to several herpesviruses. The results suggest that the effects of foscarnet and H2G on primary SIVsm infection in monkeys are direct and not mediated through inhibition of a replicating herpesvirus.
Collapse
Affiliation(s)
- D. Böttiger
- Department of Virology, Karolinska Institute, Stockholm, Sweden
| | - B. Öberg
- Department of Virology, Karolinska Institute, Stockholm, Sweden
- Medivir AB, Huddinge, Sweden
| |
Collapse
|
2
|
Abstract
Az 1986-ban felfedezett emberi 6-os herpeszvírus A és B változata molekuláris tulajdonságai alapján a legősibb emberi herpeszvírus. A B változat cseppfertőzéssel terjed a tünetmentes vírusürítő felnőttekről a két év alatti kisgyermekekre, akikben alkalmilag exanthema subitum jöhet létre. A vírus a CD4+ macrophagokat, lymphocytákat fertőzi, utóbbiakban élethossziglan lappangás, időnként a nyálmirigyekben vírustermeléssel járó perzisztencia alakul ki. Felnőttkorban ez a változat csontvelő- és szervátültetések kapcsán, immunszuppresszió talaján reaktiválódik, és akár halálos szövődményeket hoz létre. Sclerosis multiplex, idült fáradtság tünetegyüttes, Hodgkin- és nem Hodgkin-lymphomák kialakulásában kofaktor. A CD+-sejteket fertőző és bennük lappangó A változat közvetlen kórokozó képessége nem ismert. A HIV-fertőzést rendkívül erősen transzaktiváljain vitroés betegekben egyaránt. Papillomavírusok által okozott daganatokban is transzaktivátor. Mindkét vírusváltozat kórokozó képessége a megváltozott citokin- és kemokinegyensúlyon alapszik. A két változat elkülönítése szerológiailag nehézkes, erre a savóból vagy a fehérvérsejtekből végzett változatspecifikus PCR alkalmas. A súlyos komplikációk kezelésére, esetleg kemoprofilaxisára ganciclovir, esetleg foscarnet és cidofovir használható.
Collapse
Affiliation(s)
- József Ongrádi
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar Közegészségtani Intézet Budapest Nagyvárad tér 4. 1089
| | - Valéria Kövesdi
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar Közegészségtani Intézet Budapest Nagyvárad tér 4. 1089
| | - G. Péter Medveczky
- 2 University of South Florida College of Medicine Department of Molecular Medicine Tampa FL
| |
Collapse
|
3
|
Tomoiu A, Gravel A, Flamand L. Mapping of human herpesvirus 6 immediate-early 2 protein transactivation domains. Virology 2006; 354:91-102. [PMID: 16884756 DOI: 10.1016/j.virol.2006.06.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2006] [Revised: 04/10/2006] [Accepted: 06/23/2006] [Indexed: 11/21/2022]
Abstract
The immediate-early 2 (IE2) protein of human herpesvirus 6 (HHV-6) is a potent transactivator of multiple cellular and viral promoters. Deletion mutants of HHV-6 variant A IE2 allowed us to map functional transactivation domains acting on complex and minimal promoter sequences. This mapping showed that both the N-terminal and C-terminal domains of IE2 are required for efficient transactivation, and that deletion of the C-terminal (1397-1466) tail of IE2 drastically reduces both transactivation and the intranuclear distribution of IE2. Moreover, we determined that the ATF/CRE binding site within the HHV-6A polymerase promoter is not required for efficient transactivation by IE2, whereas the R3 repeat region of the putative immediate-early promoter of HHV-6A is responsive to and positively regulated by IE2. These results contrast sharply to that of human cytomegalovirus (HCMV) IE2, which down-regulates its promoter. Our characterization of HHV-6 IE2 transactivating activity provides a better understanding of the complex interactions of this protein with the viral and cellular transcription machinery and highlights significant differences with the IE2 protein of HCMV.
Collapse
Affiliation(s)
- Andru Tomoiu
- Laboratory of Virology, Rheumatology and Immunology Research Center, Centre de Recherche du CHUL and Faculty of Medicine, Laval University, 2705 Laurier Blvd., Room T1-49, Québec, Qc, Canada G1V 4G2.
| | | | | |
Collapse
|
4
|
De Bolle L, Naesens L, De Clercq E. Update on human herpesvirus 6 biology, clinical features, and therapy. Clin Microbiol Rev 2005; 18:217-45. [PMID: 15653828 PMCID: PMC544175 DOI: 10.1128/cmr.18.1.217-245.2005] [Citation(s) in RCA: 341] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human herpesvirus 6 (HHV-6) is a betaherpesvirus that is closely related to human cytomegalovirus. It was discovered in 1986, and HHV-6 literature has expanded considerably in the past 10 years. We here present an up-to-date and complete overview of the recent developments concerning HHV-6 biological features, clinical associations, and therapeutic approaches. HHV-6 gene expression regulation and gene products have been systematically characterized, and the multiple interactions between HHV-6 and the host immune system have been explored. Moreover, the discovery of the cellular receptor for HHV-6, CD46, has shed a new light on HHV-6 cell tropism. Furthermore, the in vitro interactions between HHV-6 and other viruses, particularly human immunodeficiency virus, and their relevance for the in vivo situation are discussed, as well as the transactivating capacities of several HHV-6 proteins. The insight into the clinical spectrum of HHV-6 is still evolving and, apart from being recognized as a major pathogen in transplant recipients (as exemplified by the rising number of prospective clinical studies), its role in central nervous system disease has become increasingly apparent. Finally, we present an overview of therapeutic options for HHV-6 therapy (including modes of action and resistance mechanisms).
Collapse
Affiliation(s)
- Leen De Bolle
- Rega Institute for Medical Research, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | | | | |
Collapse
|
5
|
Lu C, Zeng Y, Huang Z, Huang L, Qian C, Tang G, Qin D. Human herpesvirus 6 activates lytic cycle replication of Kaposi's sarcoma-associated herpesvirus. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:173-83. [PMID: 15632010 PMCID: PMC1602294 DOI: 10.1016/s0002-9440(10)62242-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/01/2004] [Indexed: 10/18/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) or human herpesvirus 8 (HHV-8) is a gamma-herpesvirus consistently identified in Kaposi's sarcoma (KS), primary effusion lymphoma, and multicentric Castleman's disease. KSHV infection appears to be necessary, but not be sufficient for development of KS without other co-factors. However, factors that facilitate KSHV to cause KS have not been well defined. Because patients with KS are often immunosuppressed and susceptible to many infectious agents including human herpesvirus 6 (HHV-6), we investigated the potential of HHV-6 to influence the replication of KSHV. By co-culturing HHV-6-infected T cells with KSHV-latent BCBL-1 cell line, infecting BCBL-1 cells with HHV-6 virions, and generating heterokaryons between HHV-6-infected T cells and BCBL-1 cells, we showed that HHV-6 played a critical role in induction of KSHV replication, as determined by production of lytic phase mRNA transcripts and viral proteins. We confirmed and extended the results by using a luciferase reporter assay in which KSHV ORF50 promoter, the first promoter activated during KSHV replication, drove the luciferase expression. Besides HHV-6, we also found that cytokines such as interferon-gamma partially contributed to induction of KSHV replication in the co-culture system. These findings suggest that HHV-6 may participate in KS pathogenesis by promoting KSHV replication and increasing KSHV viral load.
Collapse
Affiliation(s)
- Chun Lu
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing 210029, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
6
|
Kondo K, Sashihara J, Shimada K, Takemoto M, Amo K, Miyagawa H, Yamanishi K. Recognition of a novel stage of betaherpesvirus latency in human herpesvirus 6. J Virol 2003; 77:2258-64. [PMID: 12525662 PMCID: PMC140895 DOI: 10.1128/jvi.77.3.2258-2264.2003] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2002] [Accepted: 10/24/2002] [Indexed: 11/20/2022] Open
Abstract
Latency-associated transcripts of human herpesvirus 6 (H6LTs) (K. Kondo et al. J. Virol. 76:4145-4151, 2002) were maximally expressed at a fairly stable intermediate stage between latency and reactivation both in vivo and in vitro. H6LTs functioned as sources of immediate-early protein 1 at this stage, which up-regulated the viral reactivation.
Collapse
Affiliation(s)
- Kazuhiro Kondo
- Department of Microbiology. Department of Pediatrics, Osaka University Medical School, Suita City, Osaka 565-0871, Japan.
| | | | | | | | | | | | | |
Collapse
|
7
|
Csoma E, Bácsi A, Liu X, Szabó J, Ebbesen P, Beck Z, Kónya J, Andirkó I, Nagy E, Tóth FD. Human herpesvirus 6 variant a infects human term syncytiotrophoblasts in vitro and induces replication of human immunodeficiency virus type 1 in dually infected cells. J Med Virol 2002; 67:67-87. [PMID: 11920820 DOI: 10.1002/jmv.2194] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Human herpesvirus 6 (HHV-6) and human immunodeficiency virus type 1 (HIV-1) may interact during transplacental transmission of HIV-1. The placental syncytiotrophoblast layer serves as the first line of defense of the fetus against viruses. Patterns of replication of HHV-6 variant A (HHV-6A) and HIV-1 were analyzed in singly and dually infected human term syncytiotrophoblast cells cultured in vitro. For this purpose, the GS strain of HHV-6A and the Ba-L and IIIB strains of HIV-1 were used. HHV-6A replication was restricted at the level of early gene products in singly infected syncytiotrophoblasts, whereas no viral protein expression was found in cells infected with HIV-1 alone. Coinfection of syncytiotrophoblast cells with HHV-6A and HIV-1 resulted in production of infectious HIV-1. In contrast, no enhancement of HHV-6A expression was observed in cell cultures infected with both viruses. Uninfected syncytiotrophoblast cells were found to express CXCR4 and CCR3 but not CD4 or CCR5 receptors. Infection of syncytiotrophoblasts with HHV-6A did not induce CD4 expression and had no influence on chemokine receptor expression. Activation of HIV-1 from latency in coinfected cells was mediated by the immediate-early (IE)-A and IE-B gene products of HHV-6A. Open reading frames U86 and U89 of the IE-A region were able to activate HIV-1 replication in a synergistic manner. The data suggest that in vivo double infection of syncytiotrophoblast cells with HHV-6A and HIV-1 could contribute to the transplacental transmission of HIV-1 but not HHV-6A.
Collapse
Affiliation(s)
- Eszter Csoma
- Institute of Microbiology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Xu Y, Linde A, Dahl H, Winberg G. Definition of a divergent epitope that allows differential detection of early protein p41 from human herpesvirus 6 variants A and B. J Clin Microbiol 2001; 39:1449-55. [PMID: 11283070 PMCID: PMC87953 DOI: 10.1128/jcm.39.4.1449-1455.2001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human herpesvirus 6 (HHV-6) early protein, p41, encoded by the U27 gene has been detected in oligodendrocytes of multiple sclerosis (MS) patients by using a monoclonal antibody (MAb to p41/38). We here report the antigenic epitope of HHV-6 p41 recognized by this MAb. First, we established that the MAb to p41/38 recognizes a nuclear antigen in HHV-6A strain GS-infected cells but not in HHV-6B strain Z29-infected cells. Secondly, we compared the reactivity of the MAb to p41/38 to that of another p41-specific MAb (MAb to p41) on immunoblots with purified p41-glutathione S-transferase fusion protein from strains GS and Z29 and GS- and Z29-infected-cell lysates. The two MAbs were tested in an enzyme-linked immunosorbent assay against a panel of synthetic peptides covering the amino acid substitutions between the GS- and Z29-derived p41 proteins, as determined by DNA sequencing of our cloned isolates of the U27 gene. The MAb to p41/38 reacted specifically with a peptide comprising p41 residues 321 to 340 from strain GS. The critical residue in this peptide was serine 328, as the substitution S328N in the Z29 strain rendered the corresponding peptide nonreactive. The p41 S328 marker was present in three of three HHV-6A strains, while four of four sequenced p41 genes from HHV-6B strains had N328. Our findings are of value for the interpretation of previous findings of p41 expression in brains of MS patients and may allow a more detailed analysis of the role of HHV-6 variants in other disorders.
Collapse
Affiliation(s)
- Y Xu
- Department of Virology, Swedish Institute for Infectious Disease Control, 171 82 Solna, Sweden
| | | | | | | |
Collapse
|
9
|
Hasegawa A, Yasukawa M, Sakai I, Fujita S. Transcriptional down-regulation of CXC chemokine receptor 4 induced by impaired association of transcription regulator YY1 with c-Myc in human herpesvirus 6-infected cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:1125-31. [PMID: 11145693 DOI: 10.4049/jimmunol.166.2.1125] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have recently reported that down-regulation of CXC chemokine receptor (CXCR) 4 in CD4(+) T lymphocytes is induced by human herpesvirus (HHV) 6 infection. In this study, we further studied the mechanisms of HHV-6-induced CXCR4 down-regulation, focusing on the regulation of CXCR4 transcription. Down-regulation of CXCR4 transcription was detected in HHV-6A-infected JJHAN and HHV-6B-infected MT-4 cell lines, as we had previously reported for HHV-6-infected peripheral blood CD4(+) T lymphocytes. Luciferase assays revealed that a YY1-binding site around -320 relative to the transcription start site is important for down-regulation of CXCR4 transcription in HHV-6-infected cells. The binding activity of YY1, which is a repressor of CXCR4 transcription, to the CXCR4 promoter appeared to significantly increase in HHV-6-infected cells compared with the binding activity in mock-infected cells. Immunoprecipitation assays showed that in HHV-6-infected cells association of c-Myc with YY1 was decreased and that of Max with c-Myc was increased, whereas association of Mad with Max appeared to be decreased. The amounts of each of YY1, c-Myc, Max, and Mad proteins synthesized in cells were not altered by HHV-6 infection. These data indicate that the decreased association of YY1 with c-Myc that is caused by impaired interaction in the c-Myc/Max/Mad network results in increased binding activity of YY1 to the CXCR4 promoter, mediating down-regulation of CXCR4 production in HHV-6-infected cells.
Collapse
Affiliation(s)
- A Hasegawa
- First Department of Internal Medicine, Ehime University School of Medicine, Shigenobu, Japan
| | | | | | | |
Collapse
|
10
|
Abstract
Human herpesvirus 6 (HHV-6), a member of the beta-herpesvirinae subfamily, is highly seroprevalent, has a worldwide distribution, and infection usually occurs within the first two years of life. In this age group, HHV-6 causes febrile illness including exanthem subitum with seizures a recognised complication. The virus is predominantly T lymphotropic although it can infect a variety of cell types in vitro and CD46 has recently been identified as a cellular receptor. The virus persists in the host, with a latent state proposed in monocytes and bone marrow progenitor cells, and chronic infection in salivary glands. The virus is pathogenic in the post transplantation period and may be a cofactor in the progression of HIV disease. The virus has also been associated with multiple sclerosis (MS), with the virus detected in oligodendrocytes particularly in plaque regions. The role of HHV-6 in MS remains controversial and a more extensive understanding of its neurotropism and association with disease is required. Two variants of HHV-6 exist (A and B) and comparison of their complete nucleotide sequences shows the genomes to be colinear, with a high degree of homology. Variation in specific regions of the genome is more extensive and probably accounts for biological and pathological differences. Almost exclusively, variant B is associated with febrile illness in childhood and is the predominant variant detected in healthy individuals. The epidemiology of HHV-6A infection needs to be better defined, although it is significantly less prevalent. Biological, genetic, epidemiological and pathological findings suggest that the two variants are divergent.
Collapse
Affiliation(s)
- D A Clark
- Department of Virology, Royal Free and University College Medical School, London, UK
| |
Collapse
|
11
|
Isegawa Y, Mukai T, Nakano K, Kagawa M, Chen J, Mori Y, Sunagawa T, Kawanishi K, Sashihara J, Hata A, Zou P, Kosuge H, Yamanishi K. Comparison of the complete DNA sequences of human herpesvirus 6 variants A and B. J Virol 1999; 73:8053-63. [PMID: 10482554 PMCID: PMC112821 DOI: 10.1128/jvi.73.10.8053-8063.1999] [Citation(s) in RCA: 180] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human herpesvirus 6 (HHV-6), which belongs to the betaherpesvirus subfamily and infects mainly T cells in vitro, causes acute and latent infections. Two variants of HHV-6 have been distinguished on the basis of differences in several properties. We have determined the complete DNA sequence of HHV-6 variant B (HHV-6B) strain HST, the causative agent of exanthem subitum, and compared the sequence with that of variant A strain U1102. A total of 115 potential open reading frames (ORFs) were identified within the 161,573-bp contiguous sequence of the entire HHV-6 genome, including some genes with remarkable differences in amino acid identity. All genes with <70% identity between the two variants were found to contain deleted regions when ORFs that could not be expressed were excluded from the comparison. Except in the case of U47, these differences were found in immediate-early/regulatory genes, DR2, DR7, U86/90, U89/90, and U95, which may represent characteristic differences of variants A and B. Also, we have successfully typed 14 different strains belonging to variant A or B by PCR using variant-specific primers; the results suggest that the remarkable differences observed were conserved evolutionarily as variant-specific divergence.
Collapse
Affiliation(s)
- Y Isegawa
- Department of Microbiology, Osaka University Medical School C1, 2-2 Yamada-Oka Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Dominguez G, Dambaugh TR, Stamey FR, Dewhurst S, Inoue N, Pellett PE. Human herpesvirus 6B genome sequence: coding content and comparison with human herpesvirus 6A. J Virol 1999; 73:8040-52. [PMID: 10482553 PMCID: PMC112820 DOI: 10.1128/jvi.73.10.8040-8052.1999] [Citation(s) in RCA: 248] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/1999] [Accepted: 06/10/1999] [Indexed: 11/20/2022] Open
Abstract
Human herpesvirus 6 variants A and B (HHV-6A and HHV-6B) are closely related viruses that can be readily distinguished by comparison of restriction endonuclease profiles and nucleotide sequences. The viruses are similar with respect to genomic and genetic organization, and their genomes cross-hybridize extensively, but they differ in biological and epidemiologic features. Differences include infectivity of T-cell lines, patterns of reactivity with monoclonal antibodies, and disease associations. Here we report the complete genome sequence of HHV-6B strain Z29 [HHV-6B(Z29)], describe its genetic content, and present an analysis of the relationships between HHV-6A and HHV-6B. As sequenced, the HHV-6B(Z29) genome is 162,114 bp long and is composed of a 144,528-bp unique segment (U) bracketed by 8,793-bp direct repeats (DR). The genomic sequence allows prediction of a total of 119 unique open reading frames (ORFs), 9 of which are present only in HHV-6B. Splicing is predicted in 11 genes, resulting in the 119 ORFs composing 97 unique genes. The overall nucleotide sequence identity between HHV-6A and HHV-6B is 90%. The most divergent regions are DR and the right end of U, spanning ORFs U86 to U100. These regions have 85 and 72% nucleotide sequence identity, respectively. The amino acid sequences of 13 of the 17 ORFs at the right end of U differ by more than 10%, with the notable exception of U94, the adeno-associated virus type 2 rep homolog, which differs by only 2.4%. This region also includes putative cis-acting sequences that are likely to be involved in transcriptional regulation of the major immediate-early locus. The catalog of variant-specific genetic differences resulting from our comparison of the genome sequences adds support to previous data indicating that HHV-6A and HHV-6B are distinct herpesvirus species.
Collapse
Affiliation(s)
- G Dominguez
- Centers for Disease Control and Prevention, Atlanta, Georgia 30333, USA
| | | | | | | | | | | |
Collapse
|
13
|
Doniger J, Muralidhar S, Rosenthal LJ. Human cytomegalovirus and human herpesvirus 6 genes that transform and transactivate. Clin Microbiol Rev 1999; 12:367-82. [PMID: 10398670 PMCID: PMC100243 DOI: 10.1128/cmr.12.3.367] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This review is an update on the transforming genes of human cytomegalovirus (HCMV) and human herpesvirus 6 (HHV-6). Both viruses have been implicated in the etiology of several human cancers. In particular, HCMV has been associated with cervical carcinoma and adenocarcinomas of the prostate and colon. In vitro transformation studies have established three HCMV morphologic transforming regions (mtr), i.e., mtrI, mtrII, and mtrIII. Of these, only mtrII (UL111A) is retained and expressed in both transformed and tumor-derived cells. The transforming and tumorigenic activities of the mtrII oncogene were localized to an open reading frame (ORF) encoding a 79-amino-acid (aa) protein. Furthermore, mtrII protein bound to the tumor suppressor protein p53 and inhibited its ability to transactivate a p53-responsive promoter. In additional studies, the HCMV immediate-early protein IE86 (IE2; UL122) was found to interact with cell cycle-regulatory proteins such as p53 and Rb. However, IE86 exhibited transforming activity in vitro only in cooperation with adenovirus E1A. HHV-6 is a T-cell-tropic virus associated with AIDS-related and other lymphoid malignancies. In vitro studies identified three transforming fragments, i.e., SalI-L, ZVB70, and ZVH14. Of these, only SalI-L (DR7) was retained in transformed and tumor-derived cells. The transforming and tumorigenic activities of SalI-L have been localized to a 357-aa ORF-1 protein. The ORF-1 protein was expressed in transformed cells and, like HCMV mtrII, bound to p53 and inhibited its ability to transactivate a p53-responsive promoter. HHV-6 has also been proposed to be a cofactor in AIDS because both HHV-6 and human immunodeficiency virus type 1 (HIV-1) have been demonstrated to coinfect human CD4(+) T cells, causing accelerated cytopathic effects. Interestingly, like the transforming proteins of DNA tumor viruses such as simian virus 40 and adenovirus, ORF-1 was also a transactivator and specifically up-regulated the HIV-1 long terminal repeat when cotransfected into CD4(+) T cells. Finally, based on the interactions of HCMV and HHV-6 transforming proteins with tumor suppressor proteins, a scheme is proposed for their role in oncogenesis.
Collapse
Affiliation(s)
- J Doniger
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, D.C. 20007, USA
| | | | | |
Collapse
|
14
|
Yasukawa M, Hasegawa A, Sakai I, Ohminami H, Arai J, Kaneko S, Yakushijin Y, Maeyama K, Nakashima H, Arakaki R, Fujita S. Down-Regulation of CXCR4 by Human Herpesvirus 6 (HHV-6) and HHV-7. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.9.5417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Recent studies have demonstrated that human herpesvirus 6 (HHV-6) and HHV-7 interact with HIV-1 and alter the expression of various surface molecules and functions of T lymphocytes. The present study was undertaken to clarify whether coreceptors for HIV-1, CXCR4 and CCR5, are necessary for HHV-6 and HHV-7 infection. Although CXCR4 and CCR5 appeared not to be the coreceptors for these viruses, marked down-regulation of CXCR4, but not CCR5, was detected in HHV-6 variant A (HHV-6A)-, HHV-6 variant B (HHV-6B)-, and HHV-7-infected cells. Down-regulation of CXCR4 resulted in impairment of chemotaxis and a decreased level of elevation of the intracellular Ca2+ concentration in response to stromal cell-derived factor-1. Northern blot analysis of mRNAs extracted from HHV-6A-, HHV-6B-, and HHV-7-infected CD4+ T lymphocytes demonstrated a markedly decreased level of CXCR4 gene transcription, but the posttranscriptional stability of CXCR4 mRNA was not significantly altered. These data demonstrate that unlike HIV-1, HHV-6 and HHV-7 infections do not require expression of CXCR4 or CCR5, whereas marked down-regulation of CXCR4 is induced by these viruses, suggesting that HHV-6 and HHV-7 infections may render CD4+ T lymphocytes resistant to T lymphocyte-tropic HIV-1 infection.
Collapse
Affiliation(s)
| | | | | | | | - Junko Arai
- *First Department of Internal Medicine and
| | | | | | - Kazutaka Maeyama
- †Department of Pharmacology, Ehime University School of Medicine, Shigenobu, Ehime, Japan; and
| | - Hideki Nakashima
- ‡Department of Microbiology, Kagoshima University School of Dentistry, Kagoshima City, Kagoshima, Japan
| | - Rieko Arakaki
- ‡Department of Microbiology, Kagoshima University School of Dentistry, Kagoshima City, Kagoshima, Japan
| | | |
Collapse
|
15
|
Mori Y, Yagi H, Shimamoto T, Isegawa Y, Sunagawa T, Inagi R, Kondo K, Tano Y, Yamanishi K. Analysis of human herpesvirus 6 U3 gene, which is a positional homolog of human cytomegalovirus UL 24 gene. Virology 1998; 249:129-39. [PMID: 9740784 DOI: 10.1006/viro.1998.9305] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The US22 gene family was first discovered in human cytomegalovirus (HCMV) and contains several conserved amino acid motifs. Human herpesvirus 6 (HHV-6) also encodes several genes belonging to the US22 family, including the U3 gene (a positional homolog of HCMV UL24). Because the gene products of the US22 gene family function as gene regulators in general, we analyzed the HHV-6A U3 gene. Six transcripts with different molecular weights of 7.5-1.8 kb were detected by Northern blot analysis using a U3-specific probe. Sequence analyses of the respective cDNA clones and primer extension experiments revealed that the U3 gene encoded the 2.0- and 3.5-kb transcripts and had no splicing within the U3 gene region. By immunofluorescence testing using antibodies raised to a fusion protein of MBP (maltose-binding protein) and U3, the U3 viral antigen was detected as early as 24 h p.i. in HHV-6A-infected U373 cells. The antigens were found in cytoplasmic granules, preferentially in the endoplasmic reticulum. Moreover, cotransfection assay using a luciferase gene expression system revealed that the U3 gene product was capable of activating the human immunodeficiency virus long terminal repeat promoter in CV1 cells.
Collapse
Affiliation(s)
- Y Mori
- Departments of Microbiology, Ophthalmology, Osaka University Medical School, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Boccuni MC, Campanini F, Battista MC, Bergamini G, Dal Monte P, Ripalti A, Landini MP. Human cytomegalovirus product UL44 downregulates the transactivation of HIV-1 long terminal repeat. AIDS 1998; 12:365-72. [PMID: 9520165 DOI: 10.1097/00002030-199804000-00004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Human cytomegalovirus (HCMV) is often isolated from HIV-1-infected patients and the two viruses can infect the same cell type giving rise to direct bidirectional interactions. Whereas the long terminal repeat (LTR) transactivation ability of HCMV immediate early gene (IE1/IE2) is well documented, no information is available on the possible role of other HCMV proteins. In this study, the activity of ppUL44, an early DNA-binding protein, on HIV LTR transactivation was investigated. METHODS HIV LTR transactivation by ppUL44 in presence or absence of HIV-1 Tat and HCMV IE1/IE2 was determined in J-Jhan and U973 cells through transient transfection experiments with a series of different expression vectors. Some experiments were also performed on U373-MG astrocytoma cells permanently transfected with UL44 or with another HCMV gene used as a control (UL55). RESULTS The basal transactivation activity of the HIV LTR was not influenced by the presence of ppUL44. On the contrary, the transactivation observed in the presence of Tat, IE1/IE2 or both factors in synergy was strongly downregulated by ppUL44 in a dose-dependent manner. Deletion constructs of ppUL44 demonstrated that the region of the molecule responsible for the inhibition of the LTR is located within the last 114 amino acids at the carboxyl-terminal region. CONCLUSIONS The results obtained indicate that within the last 114 amino acids of ppUL44 there is a domain that has a negative effect on the ability of HIV-1 LTR to be activated by both its autologous transactivator Tat and the heterologous transactivator HCMV IE1/IE2 functioning individually or synergistically.
Collapse
Affiliation(s)
- M C Boccuni
- Department of Clinical and Experimental Medicine, University of Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
17
|
Chan SR, Bloomer C, Chandran B. Identification and characterization of human herpesvirus-8 lytic cycle-associated ORF 59 protein and the encoding cDNA by monoclonal antibody. Virology 1998; 240:118-26. [PMID: 9448696 DOI: 10.1006/viro.1997.8911] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Monoclonal antibody (Mab) 11D1 specific for HHV-8 showed a predominantly nuclear membrane fluorescence with about 30% of phorbol ester (TPA)-induced HHV-8-carrying BCBL-1 cells and with 2-8% of uninduced cells, but not with other herpes viruses infected cells. This Mab immunoprecipitated a 50-kDa polypeptide from BCBL-1 cells. The synthesis of this polypeptide was reduced but not inhibited by phosphonoacetic acid (PAA). A 2.3-kb cDNA insert from a cDNA library of TPA-induced BCBL-1 cells was identified by Mab 11D1. Sequence analysis shows that this cDNA is open at the 5' end and encodes two ORFs of 396AA (5' end) and 357AA (3' end). These ORFs are identical to the published HHV-8 ORFs 59 and 58, respectively in vitro transcription and translation of the cDNA resulted in the synthesis of a 50-kDa polypeptide and its partial peptide map was identical to that of the 50-kDa polypeptide detected in the TPA induced BCBL-1 cells. Riboprobe made from the cDNA insert hybridized with several viral specific RNAs from BCBL-1 cells. Levels of these RNA species were reduced, but not inhibited by PAA. These characteristics are similar to other herpes viruses genes encoding the lytic cycle associated early-late class accessory proteins that are essential for viral DNA replication. This Mab 11D1 recognizing the HHV-8 lytic cycle associated ORF 59 protein will be highly useful in monitoring the lytic replicative cycle.
Collapse
Affiliation(s)
- S R Chan
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City 66160, USA
| | | | | |
Collapse
|
18
|
Kimberlin DW. Human herpesviruses 6 and 7: identification of newly recognized viral pathogens and their association with human disease. Pediatr Infect Dis J 1998; 17:59-67; quiz 68. [PMID: 9469397 DOI: 10.1097/00006454-199801000-00013] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- D W Kimberlin
- Division of Clinical Virology, The University of Alabama at Birmingham, USA.
| |
Collapse
|
19
|
Abstract
Human herpesvirus 6 variant A (HHV-6A) and human herpesvirus 6 variant B (HHV-6B) are two closely related yet distinct viruses. These visuses belong to the Roseolovirus genus of the betaherpesvirus subfamily; they are most closely related to human herpesvirus 7 and then to human cytomegalovirus. Over 95% of people older than 2 years of age are seropositive for either or both HHV-6 variants, and current serologic methods are incapable of discriminating infection with one variant from infection with the other. HHV-6A has not been etiologically linked to any human disease, but such an association will probably be found soon. HHV-6B is the etiologic agent of the common childhood illness exanthem subitum (roseola infantum or sixth disease) and related febrile illnesses. These viruses are frequently active and associated with illness in immunocompromised patients and may play a role in the etiology of Hodgkin's disease and other malignancies. HHV-6 is a commensal inhabitant of brains; various neurologic manifestations, including convulsions and encephalitis, can occur during primary HHV-6 infection or in immunocompromised patients. HHV-6 and distribution in the central nervous system are altered in patients with multiple sclerosis; the significance of this is under investigation.
Collapse
Affiliation(s)
- D K Braun
- Eli Lilly, Lilly Corporate Center, Indianapolis, Indiana 46285, USA
| | | | | |
Collapse
|
20
|
Abstract
Human herpesvirus 6 (HHV-6) is a lymphotropic herpesvirus, and in vitro, it can productively infect many of the same cell types that human immunodeficiency virus (HIV) infects. Simultaneous infection of T cells by HIV and HHV-6 can lead to both activation of the HIV promoter and acceleration of the cytopathic effects. Several HHV-6 genes have been demonstrated to activate HIV promoter expression. Among them is a cDNA clone, pCD41 (U27), which codes for the HHV-6 DNA polymerase accessory protein. We have now further characterized the transcription pattern in the pCD41 locus and identified at least six RNA species, ranging in size from 1.2 to 4.5 kb. Northern (RNA) blot analyses showed no significant difference in RNA patterns between the HHV-6 variant A (GS) and variant B (Z29) viruses. All the RNA species detected by pCD41 are polyadenylated and polyribosome associated, suggesting that they may be actively engaged in protein synthesis. Cycloheximide and phosphonoacetic acid inhibition assay results indicate that all the pCD41 RNA species belong to the herpesviral early-late family. Using primer extension and S1 mapping techniques, the 5' and 3' ends of each transcript were mapped to different positions, and no splicing was observed.
Collapse
Affiliation(s)
- Y Zhou
- Department of Microbiology, University of Miami School of Medicine, Florida 33101, USA
| | | | | |
Collapse
|
21
|
Abstract
The mechanisms of cell death in CD4+ T cells mediated by human herpesvirus 6 (HHV-6) were investigated. The frequency of cell death in the human CD4+ T-cell line JJHAN, which had been inoculated with HHV-6 variant A or B, appeared to be augmented by tumor necrosis factor alpha (TNF-alpha). Agarose gel electrophoresis of DNA from HHV-6-inoculated cells showed DNA fragmentation in multiples of the oligonucleosome length unit. The degree of DNA fragmentation increased when HHV-6-inoculated cells were cultured in the presence of TNF-alpha. Flow cytometry and Scatchard analysis of TNF receptors revealed an increase in the number of the p55 form of TNF receptors on JJHAN cells after HHV-6 inoculation. It also appeared that treatment with anti-Fas monoclonal antibody (MAb) induced marked apoptosis in HHV-6-inoculated cells. Transmission electron microscopy showed characteristics of apoptosis, such as chromatin condensation and fragmentation of nuclei, but virus particles were hardly detected in apoptotic cells. Two-color flow cytometric analysis using anti-HHV-6 MAb and propidium iodide revealed that DNA fragmentation was present predominantly in uninfected cells but not in productively HHV-6-infected cells. In addition, JJHAN cells incubated with UV light-irradiated and ultracentrifuged culture supernatant of HHV-6-infected cells appeared to undergo apoptosis. The present study demonstrated that both HHV-6 variants A and B induce apoptosis in CD4+ T cells by indirect mechanisms, as reported recently in human immunodeficiency virus type 1 infection.
Collapse
Affiliation(s)
- Y Inoue
- First Department of Internal Medicine, Ehime University School of Medicine, Shigenobu, Japan
| | | | | |
Collapse
|
22
|
He J, McCarthy M, Zhou Y, Chandran B, Wood C. Infection of primary human fetal astrocytes by human herpesvirus 6. J Virol 1996; 70:1296-300. [PMID: 8551599 PMCID: PMC189947 DOI: 10.1128/jvi.70.2.1296-1300.1996] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Human herpesvirus 6 (HHV-6) is a lymphotropic betaherpesvirus which productively infects human CD4+ T cells and monocytes. HHV-6 is the etiologic agent for exanthem subitum (roseola), and it is well-known that central nervous system complications occur frequently during the course of HHV-6-associated disease. In addition, HHV-6 has been associated with encephalitis or encephalopathy. However, very little is known about its tropism for neural cells. There are reports that HHV-6 may infect some glial cell lines, but whether it can infect any primary neural cells is not known. Our studies show that both HHV-6A (GS) and HHV-6B (Z-29) can infect highly purified primary fetal astrocytes in vitro. Infected cells showed cytopathic effects, forming giant syncytia. In dual immunofluorescence assays, the infected cells were detected by antibodies against the HHV-6 p41 nuclear antigen and glial fibrillary acidic protein, indicating that the infected cells are indeed astrocytes. PCR and Northern (RNA) blot analyses also confirmed that the astrocytes are infected by HHV-6. The progeny virus did not alter its host range and could reinfect T cells as well as primary astrocytes. These findings suggest that infection of primary human astrocytes may play a role in the neuropathogenesis of HHV-6.
Collapse
Affiliation(s)
- J He
- Department of Microbiology and Immunology, University of Miami School of Medicine, Florida 33101, USA
| | | | | | | | | |
Collapse
|