1
|
Liu Y, Nonnemacher MR, Alexaki A, Pirrone V, Banerjee A, Li L, Kilareski E, Wigdahl B. Functional Studies of CCAAT/Enhancer Binding Protein Site Located Downstream of the Transcriptional Start Site. Clin Med Insights Pathol 2017; 10:1179555717694556. [PMID: 29162980 PMCID: PMC5692137 DOI: 10.1177/1179555717694556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/20/2016] [Indexed: 12/13/2022] Open
Abstract
Previous studies have identified a CCAAT/enhancer binding protein (C/EBP) site located downstream of the transcriptional start site (DS3). The role of the DS3 element with respect to HIV-1 transactivation by Tat and viral replication has not been characterized. We have demonstrated that DS3 was a functional C/EBPβ binding site and mutation of this site to the C/EBP knockout DS3-9C variant showed lower HIV-1 long terminal repeat (LTR) transactivation by C/EBPβ. However, it was able to exhibit similar or even higher transcription levels by Tat compared to the parental LTR. C/EBPβ and Tat together further enhanced the transcription level of the parental LAI-LTR and DS3-9C LTR, with higher levels in the DS3-9C LTR. HIV molecular clone viruses carrying the DS3-9C variant LTR demonstrated a decreased replication capacity and delayed rate of replication. These results suggest that DS3 plays a role in virus transcriptional initiation and provides new insight into C/EBP regulation of HIV-1.
Collapse
Affiliation(s)
- Yujie Liu
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Aikaterini Alexaki
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Vanessa Pirrone
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Anupam Banerjee
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Luna Li
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Evelyn Kilareski
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Abstract
Numerous viral vectors have been developed for the delivery of transgenes to specific target cells. For persistent transgene expression, vectors based on retroviruses are attractive delivery vehicles because of their ability to stably integrate their DNA into the host cell genome. Initially, vectors based on simple retroviruses were the vector of choice for such applications. However, these vectors can only transduce actively dividing cells. Therefore, much interest has turned to retroviral vectors based on the lentivirus genus because of their ability to transduce both dividing and non-dividing cells. The best characterized lentiviral vectors are derived from the human immunodeficiency virus type 1 (HIV-1). This chapter describes the basic features of the HIV-1 replication cycle and the many improvements reported for the lentiviral vector systems to increase the safety and efficiency. We also provide practical information on the production of HIV-1 derived lentiviral vectors, the cell transduction protocol and a method to determine the transduction titers of a lentiviral vector.
Collapse
Affiliation(s)
- Ying Poi Liu
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
3
|
Ka WH, Jeong YY, You JC. Identification of the HIV-1 packaging RNA sequence (Ψ) as a major determinant for the translation inhibition conferred by the HIV-1 5' UTR. Biochem Biophys Res Commun 2011; 417:501-7. [PMID: 22166215 DOI: 10.1016/j.bbrc.2011.11.149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 11/29/2011] [Indexed: 10/14/2022]
Abstract
The HIV-1 5' untranslated region (UTR) contains conserved sequences and unique structural motifs associated with many steps in virus replication. Because unspliced HIV mRNA containing the full-length UTR serves as a template for replication and transcription as well as packaging genomic RNA into virion, it has been postulated that the UTR may play a role in translational regulation. However, the effect and the region(s) responsible for translation control remain controversial. We used deletion mutations of the 5' UTR region in both cell-based and in vitro assays to determine if secondary structural elements within the 5' UTR confer translation inhibition, and to identify which of these elements are involved. The results indicate clearly that the entire HIV-1 5' UTR confers translation inhibition in vitro and in cells; the Psi (Ψ) region specifically has the most translation inhibitory activity among the highly-structured elements in the HIV-1 5' UTR. Moreover, it was found that the SL4 structure in the Psi (Ψ) region is the major determinant of translation inhibition, and that elimination of the SL4 RNA sequence led to increased translation. The results suggest a functional role for the Psi element and the SL4 structure in the translational control of HIV-1 full-length mRNA.
Collapse
Affiliation(s)
- Won Hye Ka
- Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul, South Korea
| | | | | |
Collapse
|
4
|
González OA, Ebersole JL, Huang CB. Oral infectious diseases: a potential risk factor for HIV virus recrudescence? Oral Dis 2009; 15:313-27. [PMID: 19364391 DOI: 10.1111/j.1601-0825.2009.01533.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
As the highly active antiretroviral therapy (HAART) has transitioned human immunodeficiency virus (HIV) infection into a 'chronic disease' management strategy, there is growing evidence that infection with non-HIV pathogens in HIV+ patients may have important public health implications in undermining HAART success and acquired immunodeficiency syndrome progression. Several bacterial and host cell products during infections with non-HIV pathogens have shown the capacity to regulate HIV replication in latently infected cells. A high prevalence of oral infections caused by bacteria, viruses and fungi has been described in HIV+ patients, including periodontal disease. The oral cavity appears to be a site of HIV pathogenesis and potential reservoir for the disease as HIV RNA and DNA forms are present in saliva as well as in gingival crevicular fluid, and oral epithelial cells are susceptible to either cell free or cell-associated HIV infection. The clinical and biological bases of potential associations between chronic oral inflammatory disorders, such as periodontal disease, and exacerbation of HIV viraemia have received little attention. This review attempts to evaluate the current understanding of HIV reactivation as a result of co-infection and/or inflammation induced by non-HIV pathogens in HIV-infected patients, and presents a hypothetic model about the potential role of periodontitis as a global oral infection that potentially contributes to HIV recrudescence.
Collapse
Affiliation(s)
- O A González
- Center for Oral Health Research, College of Dentistry,University of Kentucky, Lexington, KY 40536, USA
| | | | | |
Collapse
|
5
|
Rohr O, Marban C, Aunis D, Schaeffer E. Regulation of HIV-1 gene transcription: from lymphocytes to microglial cells. J Leukoc Biol 2003; 74:736-49. [PMID: 12960235 DOI: 10.1189/jlb.0403180] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Transcription is a crucial step for human immunodeficiency virus type 1 (HIV-1) expression in all infected host cells, from T lymphocytes, thymocytes, monocytes, macrophages, and dendritic cells in the immune system up to microglial cells in the central nervous system. To maximize its replication, HIV-1 adapts transcription of its integrated proviral genome by ideally exploiting the specific cellular environment and by forcing cellular stimulatory events and impairing transcriptional inhibition. Multiple cell type-specific interplays between cellular and viral factors perform the challenge for the virus to leave latency and actively replicate in a great diversity of cells, despite the variability of its long terminal repeat region in different HIV strains. Knowledge about the molecular mechanisms underlying transcriptional regulatory events helps in the search for therapeutic agents that target the step of transcription in anti-HIV strategies.
Collapse
Affiliation(s)
- Olivier Rohr
- Institut National de la Santé Recherche Médicale Unité, Strasbourg, France
| | | | | | | |
Collapse
|
6
|
Richman L, Meylan PRA, Munoz M, Pinaud S, Mirkovitch J. An adenovirus-based fluorescent reporter vector to identify and isolate HIV-infected cells. J Virol Methods 2002; 99:9-21. [PMID: 11684299 DOI: 10.1016/s0166-0934(01)00375-5] [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: 11/27/2022]
Abstract
A procedure is described that allows the simple identification and sorting of live human cells that transcribe actively the HIV virus, based on the detection of GFP fluorescence in cells. Using adenoviral vectors for gene transfer, an expression cassette including the HIV-1 LTR driving the reporter gene GFP was introduced into cells that expressed stably either the Tat transcriptional activator, or an inactive mutant of Tat. Both northern and fluorescence-activated cell sorting (FACS) analysis indicate that cells containing the functional Tat protein presented levels of GFP mRNA and GFP fluorescence several orders of magnitude higher than control cells. Correspondingly, cells infected with HIV-1 showed similar enhanced reporter gene activation. HIV-1-infected cells of the lymphocytic line Jurkat were easily identified by fluorescence-activated cell sorting (FACS) as they displayed a much higher green fluorescence after transduction with the reporter adenoviral vector. This procedure could also be applied on primary human cells as blood monocyte-derived macrophages exposed to the adenoviral LTR-GFP reporter presented a much higher fluorescence when infected with HIV-1 compared with HIV-uninfected cells. The vector described has the advantages of labelling cells independently of their proliferation status and that analysis can be carried on intact cells which can be isolated subsequently by fluorescence-activated cell sorting (FACS) for further culture. This work suggests that adenoviral vectors carrying a virus-specific transcriptional control element controlling the expressions of a fluorescent protein will be useful in the identification and isolation of cells transcribing actively the viral template, and to be of use for drug screening and susceptibility assays.
Collapse
Affiliation(s)
- Larry Richman
- Swiss Institute for Experimental Cancer Research (ISREC), Chemin des Boveresses 155, CH-1066, Epalinges, Switzerland
| | | | | | | | | |
Collapse
|
7
|
Dumais N, Bounou S, Olivier M, Tremblay MJ. Prostaglandin E(2)-mediated activation of HIV-1 long terminal repeat transcription in human T cells necessitates CCAAT/enhancer binding protein (C/EBP) binding sites in addition to cooperative interactions between C/EBPbeta and cyclic adenosine 5'-monophosphate response element binding protein. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:274-82. [PMID: 11751971 DOI: 10.4049/jimmunol.168.1.274] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous work indicates that treatment of human T cells with PGE(2) results in an increase of HIV-1 long terminal repeat (LTR) transcriptional activity. The noticed PGE(2)-mediated activation of virus gene activity required the participation of specific intracellular second messengers such as calcium and two transcription factors, i.e., NF-kappaB and CREB. We report in this work that the nuclear transcription factor CCAAT/enhancer binding protein (C/EBP) is also important for PGE(2)-dependent up-regulation of HIV-1 LTR-driven gene activity. The implication of C/EBP was shown by using a trans-dominant negative inhibitor of C/EBP (i.e., liver-enriched transcriptional inhibitory protein) and several molecular constructs carrying site-directed mutations in the C/EBP binding sites located within the HIV-1 LTR. Mutated HIV-1 LTR constructs also revealed the involvement of the two most proximal C/EBP binding sites. Data from cotransfection experiments with vectors coding for dominant negative mutants and gel mobility shift assays indicated that PGE(2)-mediated induction of HIV-1 LTR activity results from a cooperative interaction between C/EBPbeta and CREB, two members of the basic leucine zipper family of transcription factors. Altogether these findings indicate that treatment of human T cells with PGE(2) induces HIV-1 LTR activity through a complex interplay between C/EBPbeta and CREB. Such a combinatorial regulation may represent a mechanism that permits a fine regulation of HIV-1 expression by PGE(2) in human T cells.
Collapse
Affiliation(s)
- Nancy Dumais
- Centre de Recherche en Infectiologie, Hôpital CHUL, Centre Hospitalier Universitaire de Québec, Ste-Foy, Québec, Canada G1V 4G2
| | | | | | | |
Collapse
|
8
|
Kretz-Remy C, Munsch B, Arrigo AP. NFkappa B-dependent transcriptional activation during heat shock recovery. Thermolability of the NF-kappaB.Ikappa B complex. J Biol Chem 2001; 276:43723-33. [PMID: 11559696 DOI: 10.1074/jbc.m010821200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Heat shock induces the accumulation of misfolded proteins and results in the preferential expression of heat shock proteins, which help the cell to recover from thermal damage. Heat shock is a well known transcriptional activator of the human immunodeficiency virus type 1 long terminal repeat (LTR). We report here that mutations or deletions of the LTR kappaB sites impaired the LTR transcriptional activation by heat shock. Further analysis revealed that, during heat shock recovery, the NF-kappaB p65 and p50 subunits migrated into the nucleus of HeLa cells, bound to DNA, and induced kappaB-dependent reporter gene expression. This NF-kappaB activation did not depend on new transcriptional and/or translational events and on the pro-oxidant state generated by heat shock. It was not concomitant with IkappaBalpha phosphorylation and was not abolished by the expression of IkappaB kinase or IkappaBalpha dominant-negative mutants. Moreover, NF-kappaB activation and migration into the nucleus were not concomitant with IkappaBalpha/beta or p105 degradation. However, during heat shock recovery, NF-kappaB was dissociated from its complexing partners, allowing its migration into the nucleus. Hence, we describe here a novel mechanism for activation of NF-kappaB based on the thermolability of the NF-kappaB.IkappaB complex.
Collapse
Affiliation(s)
- C Kretz-Remy
- Laboratoire Stress Oxydant, Chaperons, et Apoptose, Centre de Génétique Moléculaire et Cellulaire, CNRS-UMR 5534, Université Claude Bernard Lyon I, F-69622 Villeurbanne Cedex, France
| | | | | |
Collapse
|
9
|
Dong Q, Kelkar S, Xiao Y, Joshi-Barve S, McClain CJ, Barve SS. Ethanol enhances TNF-alpha-inducible NFkappaB activation and HIV-1-LTR transcription in CD4+ Jurkat T lymphocytes. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 2000; 136:333-43. [PMID: 11079460 DOI: 10.1067/mlc.2000.110104] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
During the latent phase of human immunodeficiency virus type 1 (HIV-1) infection, CD4+ T cells carrying replication-competent proviral HIV-1 DNA play an important role in persistence of the virus. Several cofactors can induce and or amplify HIV-1 replication and negatively affect disease progression and pathogenesis. Ethanol consumption is an important risk factor for HIV-1 infection, and it has been implicated in increased HIV-1 replication and progression of infection. Because tumor necrosis factor-alpha (TNF-alpha) is an important modulator of HIV-1 replication, in the present study we examined the possible effects of ethanol on TNF-alpha-inducible signaling associated with HIV-1 replication in human CD4+ T cells (Jurkat E6-1). We demonstrate that clinically relevant ethanol concentrations significantly potentiate TNF-alpha-inducible NFkappaB. Although ethanol effectively collaborated with TNF-alpha, by itself it did not have a direct effect on NFkappaB activation. The ethanol-dependent potentiation of TNF-alpha-inducible NFkappaB nuclear translocation was observed to involve the enhanced degradation of IkappaBalpha. Additionally, the ethanol-mediated potentiation of TNF-alpha-inducible NFkappaB activation was abrogated by the known antioxidant pyrrolidinedithiocarbamate, suggesting an important mechanistic role for reactive oxygen species in this process. In correspondence with its effect on NFkappaB, ethanol was also observed to significantly enhance HIV-1 long terminal repeat-dependent transcription induced by TNF-alpha. Overall, the data provide a molecular basis for the possible role of ethanol as a cofactor that can adversely affect HIV-1 infection and pathogenesis.
Collapse
Affiliation(s)
- Q Dong
- Graduate Center for Toxicology, and Department of Internal Medicine, University of Kentucky Medical Center, Lexington 40536-0084, USA
| | | | | | | | | | | |
Collapse
|
10
|
Calzado MA, MacHo A, Lucena C, Muñoz E. Hydroxyurea inhibits the transactivation of the HIV-long-terminal repeat (LTR) promoter. Clin Exp Immunol 2000; 120:317-23. [PMID: 10792382 PMCID: PMC1905657 DOI: 10.1046/j.1365-2249.2000.01203.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
HIV-1 gene expression is regulated by the promoter/enhancer located within the U3 region of the proviral 5' LTR that contains multiple potential cis-acting regulatory sites. Here we describe that the inhibitor of the cellular ribonucleoside reductase, hydroxyurea (HU), inhibited phorbol myristate acetate- or tumour necrosis factor-alpha-induced HIV-1-LTR transactivation in both lymphoid and non-lymphoid cells in a dose-dependent manner within the first 6 h of treatment, with a 50% inhibitory concentration of 0.5 mM. This inhibition was found to be specific for the HIV-1-LTR since transactivation of either an AP-1-dependent promoter or the CD69 gene promoter was not affected by the presence of HU. Moreover, gel-shift assays in 5.1 cells showed that HU prevented the binding of the NF-kappaB to the kappaB sites located in the HIV-1-LTR region, but it did not affect the binding of both the AP-1 and the Sp-1 transcription factors. By Western blots and cell cycle analyses we detected that HU induced a rapid dephosphorylation of the pRB, the product of the retinoblastoma tumour suppressor gene, and the cell cycle arrest was evident after 24 h of treatment. Thus, HU inhibits HIV-1 promoter activity by a novel pathway that implies an inhibition of the NF-kappaB binding to the LTR promoter. The present study suggests that HU may be useful as a potential therapeutic approach for inhibition of HIV-1 replication through different pathways.
Collapse
Affiliation(s)
- M A Calzado
- Departamento de Biología Celular, Fisiología e Inmunología, Facultad de Medicina, Universidad de Córdoba, Córdoba, Spain
| | | | | | | |
Collapse
|
11
|
Rohr O, Schwartz C, Hery C, Aunis D, Tardieu M, Schaeffer E. The nuclear receptor chicken ovalbumin upstream promoter transcription factor interacts with HIV-1 Tat and stimulates viral replication in human microglial cells. J Biol Chem 2000; 275:2654-60. [PMID: 10644726 DOI: 10.1074/jbc.275.4.2654] [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: 11/06/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) infects the central nervous system and plays a direct role in the pathogenesis of AIDS dementia. However, the molecular mechanisms underlying HIV-1 expression in the central nervous system are poorly understood. We have recently reported that the nuclear receptor chicken ovalbumin upstream promoter transcription factor (COUP-TF), an orphan member of the nuclear receptor superfamily, is an activator of HIV-1 gene transcription. Here, our results show that COUP-TF stimulates HIV-1 transcription in primary cultured human microglial cells, the primary target for HIV-1 infection in brain. Run-on assays indicated that COUP-TF acts on the initiation step of transcription. Results from reverse transcription-polymerase chain reaction and immunocytochemistry analysis further revealed the importance of this factor by demonstrating that overexpression of COUP-TF leads to initiation of viral replication in primary HIV-infected human microglia. In addition, COUP-TF is able to physically interact and cooperate with the viral transactivator Tat. The combination of COUP-TF and Tat leads to NF-kappaB- and Sp1-independent enhanced transcriptional stimulation. In vitro binding studies showed that COUP-TF interacts with Tat through amino acids within the N-terminal DNA-binding domain of COUP-TF. Amino acids 48-72 in the basic and C-terminal regions of Tat are required for the binding of Tat to COUP-TF. These results suggest that COUP-TF is an essential transcription factor involved in HIV-1 expression in microglia and reveal a novel interplay of Tat and COUP-TF during regulation of viral expression.
Collapse
Affiliation(s)
- O Rohr
- Unité 338 INSERM, 5 rue Blaise Pascal, 67084 Strasbourg Cedex, France
| | | | | | | | | | | |
Collapse
|
12
|
Schwartz C, Catez P, Rohr O, Lecestre D, Aunis D, Schaeffer E. Functional interactions between C/EBP, Sp1, and COUP-TF regulate human immunodeficiency virus type 1 gene transcription in human brain cells. J Virol 2000; 74:65-73. [PMID: 10590092 PMCID: PMC111514 DOI: 10.1128/jvi.74.1.65-73.2000] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/1998] [Accepted: 09/21/1999] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) infects the central nervous system (CNS) and plays a direct role in the pathogenesis of AIDS dementia. However, mechanisms underlying HIV-1 gene expression in the CNS are poorly understood. The importance of CCAAT/enhancer binding proteins (C/EBP) for HIV-1 expression in cells of the immune system has been recently reported. In this study, we have examined the role and the molecular mechanisms by which proteins of the C/EBP family regulate HIV-1 gene transcription in human brain cells. We found that NF-IL6 acts as a potent activator of the long terminal repeat (LTR)-driven transcription in microglial and oligodendroglioma cells. In contrast, C/EBPgamma inhibits NF-IL6-induced activation. Consistent with previous data, our transient expression results show cell-type-specific NF-IL6-mediated transactivation. In glial cells, full activation needs the presence of the C/EBP binding sites; however, NF-IL6 is still able to function via the minimal -40/+80 region. In microglial cells, C/EBP sites are not essential, since NF-IL6 acts through the -68/+80 LTR region, containing two binding sites for the transcription factor Sp1. Moreover, we show that functional interactions between NF-IL6 and Sp1 lead to synergistic transcriptional activation of the LTR in oligodendroglioma and to mutual repression in microglial cells. We further demonstrate that NF-IL6 physically interacts with the nuclear receptor chicken ovalbumin upstream promoter transcription factor (COUP-TF), via its DNA binding domain, in vitro and in cells, which results in mutual transcriptional repression. These findings reveal how the interplay of NF-IL6 and C/EBPgamma, together with Sp1 and COUP-TF, regulates HIV-1 gene transcription in brain cells.
Collapse
Affiliation(s)
- C Schwartz
- Unité 338 INSERM, 67084 Strasbourg Cedex, France
| | | | | | | | | | | |
Collapse
|
13
|
Rohr O, Sawaya BE, Lecestre D, Aunis D, Schaeffer E. Dopamine stimulates expression of the human immunodeficiency virus type 1 via NF-kappaB in cells of the immune system. Nucleic Acids Res 1999; 27:3291-9. [PMID: 10454636 PMCID: PMC148562 DOI: 10.1093/nar/27.16.3291] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Recent studies have reported that lymphocytes produce, transport and bind dopamine present in plasma. However, the action of dopamine on HIV-1 gene expression in cells of the immune system has not yet been examined. Here, we have investigated the regulation of HIV-1 expression by dopamine in Jurkat T cells and in primary blood mononuclear cells (PBMC). HIV-1 replication was increased by dopamine, which correlated with the increased levels of HIV-1 transactivation. Our transient expression data revealed that dopamine stimulated transcription through the NF-kappaB element present in the long terminal repeat. The importance of NF-kappaB sites was confirmed by using vectors containing wild-type or mutant kappaB sites in a heterologous promoter. Consistent with the role of NF-kappaB in mediating dopamine responsiveness, the proteasome inhibitor MG132 abolished dopamine-induced transcriptional activation. We further explored the effect of dopamine in the presence of phorbol esters or tumor necrosis factor-alpha (TNF-alpha) known to activate NF-kappaB. The combination of dopamine and TNF-alpha led to a stimulation of HIV-1 transcription and replication. However, in contrast with TNF-alpha, dopamine treatment did not affect NF-kappaB DNA binding activity nor the concentrations of p50, p65 and IkappaB-alpha proteins, which suggests a distinct NF-kappaB activation mechanism. These results reveal a new link between the dopamine system, cytokine signaling pathway and regulation of gene expression via the involvement of NF-kappaB in T cells and PBMC.
Collapse
Affiliation(s)
- O Rohr
- Unité 338 INSERM, 5 rue Blaise Pascal, 67084 Strasbourg Cedex, France
| | | | | | | | | |
Collapse
|
14
|
Cassé C, Giannoni F, Nguyen VT, Dubois MF, Bensaude O. The transcriptional inhibitors, actinomycin D and alpha-amanitin, activate the HIV-1 promoter and favor phosphorylation of the RNA polymerase II C-terminal domain. J Biol Chem 1999; 274:16097-106. [PMID: 10347161 DOI: 10.1074/jbc.274.23.16097] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Actinomycin D and alpha-amanitin are commonly used to inhibit transcription. Unexpectedly, however, the transcription of the human immunodeficiency virus (HIV-1) long terminal repeats (LTR) is shown to be activated at the level of elongation, in human and murine cells exposed to these drugs, whereas the Rous sarcoma virus LTR, the human cytomegalovirus immediate early gene (CMV), and the HSP70 promoters are repressed. Activation of the HIV LTR is independent of the NFkappaB and TAR sequences and coincides with an enhanced average phosphorylation of the C-terminal domain (CTD) from the largest subunit of RNA polymerase II. Both the HIV-1 LTR activation and the bulk CTD phosphorylation enhancement are prevented by several CTD kinase inhibitors, including 5, 6-dichloro-1-beta-D-ribofuranosylbenzimidazole. The efficacies of the various compounds to block CTD phosphorylation and transcription in vivo correlate with their capacities to inhibit the CDK9/PITALRE kinase in vitro. Hence, the positive transcription elongation factor, P-TEFb, is likely to contribute to the average CTD phosphorylation in vivo and to the activation of the HIV-1 LTR induced by actinomycin D.
Collapse
Affiliation(s)
- C Cassé
- Laboratoire de Génétique Moléculaire, Ecole Normale Supérieure, 46 rue d'Ulm, 75230 Paris Cedex 05, France
| | | | | | | | | |
Collapse
|
15
|
Gazin C. ZFX transactivation of the HIV-1 LTR is cell specific and depends on core enhancer and TATA box sequences. Nucleic Acids Res 1999; 27:2156-64. [PMID: 10219088 PMCID: PMC148435 DOI: 10.1093/nar/27.10.2156] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The ZFX gene is ubiquitously transcribed and highly conserved among vertebrates. The integrity of Zfx, its murine homologue, has been shown to be important for growth during embryogenesis and sustained gamete production. Alternative splicing was shown to result in production of mRNAs coding for either ZFX804or a shorter isoform initiated downstream, ZFX575. ZFX575was previously shown to be a potent transactivator of the HLA-A11 promoter. Here, the HIV-1 LTR is also shown to be potently transactivated by ZFX575in several cell types, while ZFX804activity is found to be similar to that of ZFX575, null or intermediary according to the cell type. In all cell types, the HIV-1 TATA box sequence is a key element of transactivation, while the Sp1 or NFkappaB sites are variably required, according to the cell type. Overall, the results suggest that ZFX575and ZFX804could play a role in HIV-1 LTR induction as co-activators enhancing productive interactions between upstream transactivators and the basal transcription complexes recruited by the TATA box.
Collapse
Affiliation(s)
- C Gazin
- INSERM U462, Laboratoire associé du Comité de Paris de la Ligue Nationale Contre le Cancer, Centre Hayem, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75475 Paris cedex 10, France.
| |
Collapse
|
16
|
Cheng H, Tarnok J, Parks WP. Human immunodeficiency virus type 1 genome activation induced by human T-cell leukemia virus type 1 Tax protein is through cooperation of NF-kappaB and Tat. J Virol 1998; 72:6911-6. [PMID: 9658145 PMCID: PMC109905 DOI: 10.1128/jvi.72.8.6911-6916.1998] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
For productive replication of human immunodeficiency virus type 1 (HIV-1) in host cells, the viral genome-encoded transactivator Tat and several cellular transcription factors are required for efficient viral gene transcription. However, it remains unclear how the viral genome initiates transcription before Tat is transcribed or when Tat is at suboptimal levels. Here, we utilized the human T-cell leukemia type 1 Tax protein as a molecular tool to investigate the mechanism of viral gene transcription that initiates the early phase of infection of HIV-1. Tax alone does not significantly increase the activity of HIV-1 long terminal repeat (LTR) in T lymphocytes, but it markedly enhanced the replication of an infectious HIV-1 provirus with a truncated nef gene. This enhancement is preferentially mediated by the cooperation of Tax and Tat which is dependent on TAR and duplicated kappaB cis elements of the HIV-1 LTR as well as the NF-kappaB activation domain of Tax. Furthermore, phorbol myristate acetate and membrane-targeted HIV-1 Nef also enhanced the LTR activity in the presence of Tat in the TAR- and kappaB cis element-dependent manner. These data suggest that activated NF-kappaB can functionally interact with a suboptimal amount of Tat and the HIV-1 LTR for efficient initiation of viral gene transcription.
Collapse
MESH Headings
- Binding Sites
- Gene Expression Regulation, Viral
- Gene Products, nef/genetics
- Gene Products, nef/metabolism
- Gene Products, tat/metabolism
- Gene Products, tax/genetics
- Gene Products, tax/metabolism
- Genome, Viral
- HIV Long Terminal Repeat
- HIV-1/genetics
- HIV-1/metabolism
- HIV-1/physiology
- Human T-lymphotropic virus 1/metabolism
- Humans
- Jurkat Cells
- Mitogens/metabolism
- Mitogens/pharmacology
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Phytohemagglutinins/metabolism
- Phytohemagglutinins/pharmacology
- Tetradecanoylphorbol Acetate/metabolism
- Tetradecanoylphorbol Acetate/pharmacology
- Transcription, Genetic
- Virus Replication
- nef Gene Products, Human Immunodeficiency Virus
- tat Gene Products, Human Immunodeficiency Virus
Collapse
Affiliation(s)
- H Cheng
- Department of Microbiology and Pediatrics, New York University School of Medicine, New York, New York 10016, USA
| | | | | |
Collapse
|
17
|
Estable MC, Bell B, Hirst M, Sadowski I. Naturally occurring human immunodeficiency virus type 1 long terminal repeats have a frequently observed duplication that binds RBF-2 and represses transcription. J Virol 1998; 72:6465-74. [PMID: 9658089 PMCID: PMC109809 DOI: 10.1128/jvi.72.8.6465-6474.1998] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/1997] [Accepted: 04/15/1998] [Indexed: 02/08/2023] Open
Abstract
Approximately 38% of human immunodeficiency virus type 1 (HIV-1)-infected patients within the Vancouver Lymphadenopathy-AIDS Study have proviruses bearing partial 15- to 34-nucleotide duplications upstream of the NF-kappaB binding sites within the 5' long terminal repeat (LTR). This most frequent naturally occurring length polymorphism (MFNLP) of the HIV-1 5' LTR encompasses potential binding sites for several candidate transcription factors, including TCF-1alpha/hLEF, c-Ets, AP-4, and Ras-responsive binding factor 2 (RBF-2) (M. C. Estable et al., J. Virol. 70:4053-4062, 1996). RBF-2 and an apparently related factor, RBF-1, bind to at least four cis elements within the LTR which are required for full transcriptional responsiveness to protein-tyrosine kinases and v-Ras (B. Bell and I. Sadowski, Oncogene 13:2687-2697, 1996). Here we demonstrate that representative MFNLPs from two patients specifically bind RBF-2. In both cases, deletion of the MFNLP caused elevated LTR-directed transcription in cells expressing RBF-2 but not in cells with undetectable RBF-2. RBF-1, but not RBF-2, appears to contain the Ets transcription factor family member GABPalpha/GABPbeta1. Taken together with the fact that every MFNLP from a comparative study of over 500 LTR sequences from 42 patients contains a predicted binding site for RBF-2, our data suggest that the MFNLP is selected in vivo because it provides a duplicated RBF-2 cis element, which may limit transcription in monocytes and activated T cells.
Collapse
Affiliation(s)
- M C Estable
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, UBC Center for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
18
|
Gualberto A, Marquez G, Carballo M, Youngblood GL, Hunt SW, Baldwin AS, Sobrino F. p53 transactivation of the HIV-1 long terminal repeat is blocked by PD 144795, a calcineurin-inhibitor with anti-HIV properties. J Biol Chem 1998; 273:7088-93. [PMID: 9507019 DOI: 10.1074/jbc.273.12.7088] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous reports have indicated that benzothiophenes exhibit broad anti-inflammatory properties and inhibit human immunodeficiency virus-type 1 (HIV-1) replication. We show that the immunosuppressant cyclosporin A (CsA) and benzothiophene-2-carboxamide, 5-methoxy-3-(1-methyl ethoxy)-1-oxide (PD 144795) block the induction of p53 and NF-kappaB binding to the HIV-1 long terminal repeat (LTR) by the T cell receptor activator phytohemagglutinin. CsA and PD 144795 also inhibit the induction by phytohemagglutinin of the transcription mediated by an HIV-1 LTR fragment containing the p53 and NF-kappaB sites. These effects of PD 144795 on HIV-1 transcription correlate with its ability to inhibit the phosphatase activity of calcineurin and are similar to those previously described for CsA. Moreover, a constitutive active form of calcineurin is able to induce expression from the HIV-1 LTR in a p53- and NF-kappaB-dependent manner and PD 144795 is able to block this induction. These results demonstrate that the DNA binding of p53 to the HIV-1 LTR can be modulated by calcineurin and provide a framework to understand the anti-HIV properties of benzothiophene derivatives.
Collapse
Affiliation(s)
- A Gualberto
- Department of Physiology and Biophysics and Ireland Cancer Center, CWRU School of Medicine, Cleveland, Ohio 44106, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Kretz-Remy C, Bates EE, Arrigo AP. Amino acid analogs activate NF-kappaB through redox-dependent IkappaB-alpha degradation by the proteasome without apparent IkappaB-alpha phosphorylation. Consequence on HIV-1 long terminal repeat activation. J Biol Chem 1998; 273:3180-91. [PMID: 9452429 DOI: 10.1074/jbc.273.6.3180] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We report here that amino acid analogs, which activate hsp70 promoter, are powerful transcriptional activators of human immunodeficiency virus 1 (HIV-1) long terminal repeat (LTR), an activation which was impaired when the two kappaB sites present in the LTR were mutated or deleted. Amino acid analogs also stimulated the transcription of a kappaB-controlled reporter gene. Upon treatment with amino acid analogs, the two NF-kappaB subunits (p65 and p50), which are characterized by a relatively long half-life, redistributed into the nucleus where they bound to kappaB elements. This phenomenon, which began to be detectable after 1 h of treatment, was concomitant with the degradation of the short lived inhibitory subunit IkappaB-alpha by the proteasome. However, contrasting with other NF-kappaB inducers that trigger IkappaB-alpha degradation through a phosphorylation step, amino acid analogs did not change IkappaB-alpha isoform composition. Antioxidant conditions inhibited amino acid analog stimulatory action toward NF-kappaB. This suggests that aberrant protein conformation probably generates a pro-oxidant state that is necessary for IkappaB-alpha proteolysis by the proteasome. Moreover, this activation of NF-kappaB appeared different from that mediated by endoplasmic reticulum overload as it was not inhibited by calcium chelation.
Collapse
Affiliation(s)
- C Kretz-Remy
- Laboratoire du Stress Cellulaire, Centre de Génétique Moléculaire et Cellulaire, CNRS-UMR 5534, Université Claude Bernard Lyon-I, 69622 Villeurbanne Cedex, France
| | | | | |
Collapse
|
20
|
Rohr O, Aunis D, Schaeffer E. COUP-TF and Sp1 interact and cooperate in the transcriptional activation of the human immunodeficiency virus type 1 long terminal repeat in human microglial cells. J Biol Chem 1997; 272:31149-55. [PMID: 9388268 DOI: 10.1074/jbc.272.49.31149] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We have recently reported that chicken ovalbumin upstream promoter transcription factor (COUP-TF) activates human immunodeficiency virus type 1 (HIV-1) gene transcription in glial and neuronal cells. Here, we have examined the role of COUP-TF in microglial cells, the major target cells for HIV-1 infection in brain. We show that COUP-TF activates gene expression from both the lymphotropic LAI and the macrophage-tropic JR-FL HIV-1 strains. Although COUP-TF binds to the -352/-320 nuclear receptor responsive element of the long terminal repeat, it functions as a transcriptional activator by acting on the -68/+29 minimal promoter. This region is a direct target of transcription factors Sp1 and Sp3. We report the discovery and features of a physical and functional interplay between COUP-TF and Sp1. Our cotransfection experiments provide evidence for a functional synergism between Sp1 and COUP-TF leading to enhanced transcriptional activity of the HIV-1 long terminal repeat through the Sp1 element. In contrast, Sp3 functions as a repressor of Sp1- or COUP-TF-induced activation. We further demonstrate that COUP-TF and Sp1 are capable of physically interacting, via the DNA-binding domain of COUP-TF, in vitro and in the cell. These findings reveal how the novel interplay of Sp1 and COUP-TF families of transcription factors regulate HIV-1 gene expression.
Collapse
Affiliation(s)
- O Rohr
- Unité 338 INSERM, 5 rue Blaise Pascal, 67084 Strasbourg Cedex, France
| | | | | |
Collapse
|
21
|
Abstract
Regulation of transcription is a key feature of the HIV-1 life cycle. In recent years, various sequence elements and transcription factors have been shown to participate in HIV-1 transcription control. New evidence, however, has shown that chromatin organization plays a key role in the establishment of a transcriptionally regulated HIV-1 LTR. The present review discusses recent data obtained on reconstituted and genomic HIV-1 chromatin templates.
Collapse
Affiliation(s)
- J Mirkovitch
- Swiss Institute for Experimental Cancer Research (ISREC), Epalinges, Switzerland.
| |
Collapse
|
22
|
Yasui H, Butscher W, Cohen M, Spriggs N, Wersto R, Kohn EC, Liotta L, Gardner K. Selective inhibition of mitogen-induced transactivation of the HIV long terminal repeat by carboxyamidotriazole. Calcium influx blockade represses HIV-1 transcriptional activation. J Biol Chem 1997; 272:28762-70. [PMID: 9353347 DOI: 10.1074/jbc.272.45.28762] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Carboxyamidotriazole (CAI) is a calcium influx inhibitor that has both antiproliferative and antimetastatic activities. Pretreatment of human T-cells with micromolar concentrations of CAI causes a near complete inhibition of calcium-regulated mitogen-induced transcription from the human immunodeficiency virus (HIV) long terminal repeat (LTR). This inhibition is selective since other mitogen-activated gene regulatory elements, such as the 12-O-tetradecanoylphorbol-13-acetate response element, are not influenced by the drug. HIV LTR transcription inhibition is maximal at 1.0 microM CAI, requires a pretreatment interval of at least 8 h for optimum inhibition, and shows no acute interference with the growth properties of the cells. Moreover, the inhibition is rapidly reversible upon removal of the drug from the medium. Studies to identify enhancer elements within the HIV LTR that are functionally sensitive to low-dose long-term pretreatment with CAI indicate that the NF-kappaB-binding sites are among the major targets of drug action. In vitro DNA binding studies with nuclear extracts prepared from mitogen-induced T-cells stimulated in the presence of CAI indicate that the drug differentially influences the calcium-regulated downstream signal transduction pathways necessary for specific NF-kappaB DNA binding activity at the two kappaB sites within the HIV LTR. Studies with ionomycin and thapsigargin show that repression is specific for selected modes of inducible calcium entry and indicate that, in T-cells, a major mechanism of CAI action is to modulate calcium influx at a level that is proximal to the regulated release of calcium from intracellular stores. Measurement of calcium influx in CAI-treated cells reveals a dramatic and reversible inhibition of mitogen-induced calcium influx. These results indicate that CAI can be an important and effective pharmacological tool for analysis of the calcium-dependent modulation of HIV LTR transcription.
Collapse
Affiliation(s)
- H Yasui
- Laboratory of Pathology, NCI, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Schwartz C, Canonne-Hergaux F, Aunis D, Schaeffer E. Characterization of nuclear proteins that bind to the regulatory TGATTGGC motif in the human immunodeficiency virus type 1 long terminal repeat. Nucleic Acids Res 1997; 25:1177-84. [PMID: 9092627 PMCID: PMC146561 DOI: 10.1093/nar/25.6.1177] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We have recently elucidated the nature and function of transcription factors present in Jurkat, glial and neuronal cells that interact with modulatory region B, the nuclear receptor responsive element, in the long terminal repeat of human immunodeficiency virus type 1 (HIV-1). Considering the key role that the combination of host cell proteins plays in HIV-1 gene transcription, it appears essential to characterize proteins interacting with the adjacent region A. In vitro experiments revealed that the 5'-TGATTGGC-3'motif of region A is the target for at least three distinct proteins, one belonging to the nuclear factor I family, while two others are related to the cAMP response element binding (CREB) protein family. One of these proteins, present in DNA-protein complex C2, is formed by distinct polypeptides of relative molecular mass 43 000 and 50 000. We have purified the 43 kDa protein, which is distinct from CREB-43, and have shown that renatured p43 is able to specifically interact with site A. Transient expression experiments with vectors containing wild-type or mutant motif A revealed that basal HIV-1 gene transcription in Jurkat cells is regulated by antagonistic effects of the site A binding proteins.
Collapse
Affiliation(s)
- C Schwartz
- Unité 338 INSERM, Centre de Neurochimie, 5 rue Blaise Pascal, 67084 Strasbourg Cedex, France
| | | | | | | |
Collapse
|
24
|
Das AT, Klaver B, Klasens BI, van Wamel JL, Berkhout B. A conserved hairpin motif in the R-U5 region of the human immunodeficiency virus type 1 RNA genome is essential for replication. J Virol 1997; 71:2346-56. [PMID: 9032371 PMCID: PMC191344 DOI: 10.1128/jvi.71.3.2346-2356.1997] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The untranslated leader region of the human immunodeficiency virus (HIV) RNA genome contains multiple hairpin motifs. The repeat region of the leader, which is reiterated at the 3' end of the RNA molecule, encodes the well-known TAR hairpin and a second hairpin structure with the polyadenylation signal AAUAAA in the single-stranded loop [the poly(A) hairpin]. The fact that this poly(A) stem-loop structure and its thermodynamic stability are well conserved among HIV and simian immunodeficiency virus isolates, despite considerable divergence in sequence, suggests a biological function for this RNA motif in viral replication. Consistent with this idea, we demonstrate that mutations that alter the stability of the stem region or delete the upper part of the hairpin do severely inhibit replication of HIV type 1. Whereas destabilizing mutations in either the left- or right-hand side of the base-paired stem interfere with virus replication, the double mutant, which allows the formation of new base pairs, replicates more rapidly than the two individual virus mutants. Upon prolonged culturing of viruses with an altered hairpin stability, revertant viruses were obtained with additional mutations that restore the thermodynamic stability of the poly(A) hairpin. Transient transfection experiments demonstrated that transcription of the proviral genomes, translation of the viral mRNAs, and reverse transcription of the genomic RNAs are not affected by mutation of the 5' poly(A) hairpin. We show that the genomic RNA content of the virions is reduced by destabilization of this poly(A) hairpin but not by stabilization or truncation of this structure. These results suggest that the formation of the poly(A) hairpin structure at the 5' end of the genomic RNA molecule is necessary for packaging of viral genomes into virions and/or stability of the virion RNA.
Collapse
Affiliation(s)
- A T Das
- Department of Human Retrovirology, Academic Medical Center, University of Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
25
|
Sawaya BE, Rohr O, Aunis D, Schaeffer E. Chicken ovalbumin upstream promoter transcription factor, a transcriptional activator of HIV-1 gene expression in human brain cells. J Biol Chem 1996; 271:23572-6. [PMID: 8798567 DOI: 10.1074/jbc.271.38.23572] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Viral infection of the central nervous system by the human immunodeficiency virus type 1 leads to a wide range of neuropathological disorders. However, the molecular mechanisms governing transcription of the human immunodeficiency virus type 1 genome in brain remain unclear. We have recently established that in brain cells, proteins belonging to the steroid/thyroid/retinoic acid receptor family bind to the -352 to -320 region of the long terminal repeat (LTR). Here, by supershift experiments, we have identified chicken ovalbumin upstream promoter transcription factor (COUP-TF), an orphan member of this nuclear receptor family, as one of the major proteins interacting with this LTR site. Cotransfection studies revealed that COUP-TF is able to dramatically activate LTR-directed gene transcription in human oligodendroglioma but not in astrocytoma cells. This activation occurs through two mechanisms, depending on the LTR sequence. Moreover, in neuronal cells COUP-TF and dopamine, a catecholamine neurotransmitter, enhance LTR-directed transcription by acting on the proximal LTR region. These results reveal the importance of COUP-TF and the dopamine signaling pathway as activators of human immunodeficiency virus type 1 gene expression in brain.
Collapse
Affiliation(s)
- B E Sawaya
- Unité 338 INSERM, 5, rue Blaise Pascal, 67084 Strasbourg, France
| | | | | | | |
Collapse
|
26
|
el Kharroubi A, Martin MA. cis-acting sequences located downstream of the human immunodeficiency virus type 1 promoter affect its chromatin structure and transcriptional activity. Mol Cell Biol 1996; 16:2958-66. [PMID: 8649407 PMCID: PMC231290 DOI: 10.1128/mcb.16.6.2958] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
We have examined the roles of AP-1, AP-3-like, DBF1, and Sp1 binding sites, which are located downstream of the human immunodeficiency virus type 1 (HIV-1) promoter, in regulating basal transcriptional activity directed by the integrated viral long terminal repeat (LTR). Point mutations affecting all four of these elements functionally inactivated the HIV-1 LTR when it was constrained in a chromatin configuration. Analyses of the chromatin structures of the transcriptionally active wild-type and inactive mutated HIV-1 promoters revealed several differences. In the active promoter, the 3' half of the U3 region, including the basal promoter, the enhancer, and the putative upstream regulatory sequences are situated within a nuclease-hypersensitive region. However, the far upstream U3 region appears to be packaged into a nuclease-resistant nucleosomal structure, whereas the R, U5, and gag leader sequences are associated with a region of altered chromatin that is sensitive to restriction endonucleases. In the inactive template, only the basal promoter and enhancer element remain sensitive to nucleases, and the adjacent upstream and downstream regions are incorporated into nuclease-resistant nucleosomal structures. Taken together, these results indicate that the chromatin structure of the integrated HIV-1 LTR plays a critical role in modulating basal transcriptional activity.
Collapse
Affiliation(s)
- A el Kharroubi
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
27
|
Kinter A, Fauci AS. Interleukin-2 and human immunodeficiency virus infection: pathogenic mechanisms and potential for immunologic enhancement. Immunol Res 1996; 15:1-15. [PMID: 8739561 DOI: 10.1007/bf02918280] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
A hallmark of human immunodeficiency virus (HIV) infection is the progressive loss of CD4+ T lymphocytes; however, qualitative defects in immune responses occur prior to the precipitous drop CD4+ T cell numbers. One of the first immunologic defects to be described in HIV-infected individuals is a deficiency in interleukin (IL)-2 production. The addition of IL-2 in vitro to cultures of mononuclear cells from HIV-infected individuals partially or completely restored certain defective cellular immune responses. However, production of or addition of IL-2 has also been associated with increased viral replication in infected T cells. These observations underscore the pernicious correlation between immune activation and HIV replication. However, recent in vitro and in vivo studies have provided promising preliminary results suggesting that, at least at certain stages of disease, the benefits of IL-2 mediated immune enhancement may outweigh or override the inductive effects of this cytokine on HIV production.
Collapse
Affiliation(s)
- A Kinter
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md, USA
| | | |
Collapse
|
28
|
Gualberto A, Baldwin AS. p53 and Sp1 interact and cooperate in the tumor necrosis factor-induced transcriptional activation of the HIV-1 long terminal repeat. J Biol Chem 1995; 270:19680-3. [PMID: 7649977 DOI: 10.1074/jbc.270.34.19680] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Tumor necrosis factor alpha (TNF) is a potent activator of transcription directed by the human immunodeficiency virus type 1 (HIV-1) long terminal repeat (LTR). We have recently reported that the p53 tumor suppressor gene product binds to a site within the Sp1 binding region of the HIV-1 LTR and contributes to the TNF induction of this promoter. In this study we show that the transcription factor Sp1 cooperates with p53 in the transcriptional activation directed by the HIV-1 LTR. The presence of Sp1 increased p53 binding to its recognition sequence in the HIV-1 LTR, and experiments in Drosophila cells show that Sp1 is necessary for full transactivation by mutant p53. Importantly, TNF induced the association between p53 and Sp1 in Jurkat T cells. These data demonstrate a synergistic role for these proteins in the mechanism of TNF induction of HIV-1 LTR-mediated transcription and suggest that Sp1 may play an important role in modulating certain functions of p53.
Collapse
Affiliation(s)
- A Gualberto
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill 27599, USA
| | | |
Collapse
|