1
|
Bretscher C, Marchini A. H-1 Parvovirus as a Cancer-Killing Agent: Past, Present, and Future. Viruses 2019; 11:v11060562. [PMID: 31216641 PMCID: PMC6630270 DOI: 10.3390/v11060562] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 12/13/2022] Open
Abstract
The rat protoparvovirus H-1PV is nonpathogenic in humans, replicates preferentially in cancer cells, and has natural oncolytic and oncosuppressive activities. The virus is able to kill cancer cells by activating several cell death pathways. H-1PV-mediated cancer cell death is often immunogenic and triggers anticancer immune responses. The safety and tolerability of H-1PV treatment has been demonstrated in early clinical studies in glioma and pancreatic carcinoma patients. Virus treatment was associated with surrogate signs of efficacy including immune conversion of tumor microenvironment, effective virus distribution into the tumor bed even after systemic administration, and improved patient overall survival compared with historical control. However, monotherapeutic use of the virus was unable to eradicate tumors. Thus, further studies are needed to improve H-1PV's anticancer profile. In this review, we describe H-1PV's anticancer properties and discuss recent efforts to improve the efficacy of H-1PV and, thereby, the clinical outcome of H-1PV-based therapies.
Collapse
Affiliation(s)
- Clemens Bretscher
- Laboratory of Oncolytic Virus Immuno-Therapeutics, F011, German Cancer Research Center, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany.
| | - Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics, F011, German Cancer Research Center, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany.
- Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, 84 Val Fleuri, L-1526 Luxembourg, Luxembourg.
| |
Collapse
|
2
|
The MVMp P4 promoter is a host cell-type range determinant in vivo. Virology 2017; 506:141-151. [PMID: 28391161 DOI: 10.1016/j.virol.2017.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/15/2017] [Accepted: 03/23/2017] [Indexed: 11/24/2022]
Abstract
The protoparvovirus early promoters, e.g. P4 of Minute Virus of Mice (MVM), play a critical role during infection. Initial P4 activity depends on the host transcription machinery only. Since this is cell-type dependent, it is hypothesized that P4 is a host cell-type range determinant. Yet host range determinants have mapped mostly to capsid, never P4. Here we test the hypothesis using the mouse embryo as a model system. Disruption of the CRE element of P4 drastically decreased infection levels without altering range. However, when we swapped promoter elements of MVM P4 with those from equivalent regions of the closely related H1 virus, we observed elimination of infection in fibroblasts and chondrocytes and the acquisition of infection in skeletal muscle. We conclude that P4 is a host range determinant and a target for modifying the productive infection potential of the virus - an important consideration in adapting these viruses for oncotherapy.
Collapse
|
3
|
Angelova AL, Geletneky K, Nüesch JPF, Rommelaere J. Tumor Selectivity of Oncolytic Parvoviruses: From in vitro and Animal Models to Cancer Patients. Front Bioeng Biotechnol 2015; 3:55. [PMID: 25954743 PMCID: PMC4406089 DOI: 10.3389/fbioe.2015.00055] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/05/2015] [Indexed: 11/23/2022] Open
Abstract
Oncolytic virotherapy of cancer is among the innovative modalities being under development and especially promising for targeting tumors, which are resistant to conventional treatments. Presently, at least a dozen of viruses, belonging to nine different virus families, are being tested within the frames of various clinical studies in cancer patients. Continuously growing preclinical evidence showing that the autonomous rat parvovirus H-1 (H-1PV) is able to kill tumor cells that resist conventional treatments and to achieve a complete cure of various human tumors in animal models argues for its inclusion in the arsenal of oncolytic viruses with an especially promising bench to bedside translation potential. Oncolytic parvovirus safe administration to humans relies on the intrinsic preference of these agents for quickly proliferating, metabolically, and biochemically disturbed tumor versus normal cells (tumor selectivity or oncotropism). The present review summarizes and discusses (i) preclinical evidence of H-1PV innocuousness for normal cells and healthy tissues in vitro and in animals, respectively, (ii) toxicological assessments of H-1PV mono- or combined therapy in tumor-bearing virus-permissive animal models, as well as (iii) historical results of experimental infection of human cancer patients with H-1PV. Altogether, these data argue against a risk of H-1PV inducing significant toxic effects in human patients. This highly favorable safety profile allowed the translation of H-1PV preclinical research into a Phase I/IIa clinical trial being currently in progress.
Collapse
Affiliation(s)
- Assia L Angelova
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Karsten Geletneky
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany ; Department of Neurosurgery, University of Heidelberg , Heidelberg , Germany
| | - Jürg P F Nüesch
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Jean Rommelaere
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| |
Collapse
|
4
|
Autonomous parvoviruses neither stimulate nor are inhibited by the type I interferon response in human normal or cancer cells. J Virol 2014; 88:4932-42. [PMID: 24554651 DOI: 10.1128/jvi.03508-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Members of the genus Parvovirus are small, nonenveloped single-stranded DNA viruses that are nonpathogenic in humans but have potential utility as cancer therapeutics. Because the innate immune response to parvoviruses has received relatively little attention, we compared the response to parvoviruses to that of several other types of viruses in human cells. In normal human glia, fibroblasts, or melanocytes, vesicular stomatitis virus evoked robust beta interferon (IFN-β) responses. Cytomegalovirus, pseudorabies virus, and Sindbis virus all evoked a 2-log-unit or greater upregulation of IFN-β in glia; in contrast, LuIII and MVMp parvoviruses did not evoke a detectable IFN-β or interferon-stimulated gene (ISG; MX1, oligoadenylate synthetase [OAS], IFIT-1) response in the same cell types. The lack of response raised the question of whether parvoviral infection can be attenuated by IFN; interestingly, we found that IFN did not decrease parvovirus (MVMp, LuIII, and H-1) infectivity in normal human glia, fibroblasts, or melanocytes. The same was true in human cancers, including glioma, sarcoma, and melanoma. Similarly, IFN failed to attenuate transduction by the dependovirus vector adeno-associated virus type 2. Progeny production of parvoviruses was also unimpaired by IFN in both glioma and melanoma, whereas vesicular stomatitis virus replication was blocked. Sarcoma cells with upregulated IFN signaling that show high levels of resistance to other viruses showed strong infection by LuIII. Unlike many other oncolytic viruses, we found no evidence that impairment of innate immunity in cancer cells plays a role in the oncoselectivity of parvoviruses in human cells. Parvoviral resistance to the effects of IFN in cancer cells may constitute an advantage in the virotherapy of some tumors. IMPORTANCE Understanding the interactions between oncolytic viruses and the innate immune system will facilitate employing these viruses as therapeutic agents in cancer patients. The cancer-selective nature of some oncolytic viruses is based on the impaired innate immunity of many cancer cells. The parvoviruses H-1, LuIII, and MVM target cancer cells; however, their relationship with the innate immune system is relatively uncharacterized. Surprisingly, we found that these parvoviruses do not evoke an interferon response in normal human fibroblasts, glia, or melanocytes. Furthermore, unlike most other types of virus, we found that parvovirus infectivity is unaffected by interferon treatment of human normal or tumor cells. Finally, parvoviral replication was unimpaired by interferon in four human tumor types, including those with residual interferon functionality. We conclude that deficits in the interferon antiviral response of cancer cells do not contribute to parvoviral oncoselectivity in human cells. The interferon-resistant phenotype of parvoviruses may give them an advantage over interferon-sensitive oncolytic viruses in tumors showing residual interferon functionality.
Collapse
|
5
|
Nüesch JPF, Lacroix J, Marchini A, Rommelaere J. Molecular pathways: rodent parvoviruses--mechanisms of oncolysis and prospects for clinical cancer treatment. Clin Cancer Res 2012; 18:3516-23. [PMID: 22566376 DOI: 10.1158/1078-0432.ccr-11-2325] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Rodent parvoviruses (PV) are recognized for their intrinsic oncotropism and oncolytic activity, which contribute to their natural oncosuppressive effects. Although PV uptake occurs in most host cells, some of the subsequent steps leading to expression and amplification of the viral genome and production of progeny particles are upregulated in malignantly transformed cells. By usurping cellular processes such as DNA replication, DNA damage response, and gene expression, and/or by interfering with cellular signaling cascades involved in cytoskeleton dynamics, vesicular integrity, cell survival, and death, PVs can induce cytostasis and cytotoxicity. Although productive PV infections normally culminate in cytolysis, virus spread to neighboring cells and secondary rounds of infection, even abortive infection or the sole expression of the PV nonstructural protein NS1, is sufficient to cause significant tumor cell death, either directly or indirectly (through activation of host immune responses). This review highlights the molecular pathways involved in tumor cell targeting by PVs and in PV-induced cell death. It concludes with a discussion of the relevance of these pathways to the application of PVs in cancer therapy, linking basic knowledge of PV-host cell interactions to preclinical assessment of PV oncosuppression.
Collapse
Affiliation(s)
- Jürg P F Nüesch
- Infection and Cancer Program, Division F010, German Cancer Research Center, Heidelberg, Germany
| | | | | | | |
Collapse
|
6
|
Dempe S, Stroh-Dege AY, Schwarz E, Rommelaere J, Dinsart C. SMAD4: a predictive marker of PDAC cell permissiveness for oncolytic infection with parvovirus H-1PV. Int J Cancer 2010; 126:2914-27. [PMID: 19856310 DOI: 10.1002/ijc.24992] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents the eighth frequent solid tumor and fourth leading cause of cancer death. Because current treatments against PDAC are still unsatisfactory, new anticancer strategies are required, including oncolytic viruses. Among these, autonomous parvoviruses (PV), like MVMp (minute virus of mice) and H-1PV are being explored as candidates for cancer gene therapy. Human PDAC cell lines were identified to display various susceptibilities to an infection with H-1PV. The correlation between the integrity of the transcription factor SMAD4, mutated in 50% of all PDAC, and H-1PV permissiveness was particularly striking. Indeed, mutation or deletion of SMAD4 dramatically reduced the activity of the P4 promoter and, consequently, the accumulation of the pivotal NS1 protein. By means of DNA affinity immunoblotting, novel binding sites for SMAD4 and c-JUN transcription factors could be identified in the P4 promoter of H-1PV. The overexpression of wild-type SMAD4 in deficient cell lines (AsPC-1, Capan-1) stimulated the activity of the P4 promoter, whereas interference of endogenous SMAD4 function with a dominant-negative mutant decreased the viral promoter activity in wild-type SMAD4-expressing cells (Panc-1, MiaPaCa-2) reducing progeny virus production. In conclusion, the importance of members of the SMAD family for H-1PV early promoter P4 activity should guide us to select SMAD4-positive PDACs, which may be possible targets for an H-1PV-based cancer therapy.
Collapse
Affiliation(s)
- Sebastian Dempe
- Abt F010, Infection and Cancer Program, Tumor Virology Division, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 242, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
7
|
Augmented transgene expression in transformed cells using a parvoviral hybrid vector. Cancer Gene Ther 2008; 15:252-67. [PMID: 18202715 DOI: 10.1038/sj.cgt.7701113] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Autonomous parvoviruses possess an intrinsic oncotropism based on viral genetic elements controlling gene expression and genome replication. We constructed a hybrid vector consisting of the H1 parvovirus-derived expression cassette comprising the p4 promoter, the ns1 gene and the p38 promoter flanked by the adeno-associated viruses 2 (AAV2) inverted terminal repeats and packaged into AAV2 capsids. Gene transduction using this vector could be stimulated by coinfection with adenovirus, by irradiation or treatment with genotoxic agents, similar to standard AAV2 vectors. However, the latter were in most cases less efficient in gene transduction than the hybrid vector. With the new vector, tumor cell-selective increase in transgene expression was observed in pairs of transformed and non-transformed cells, leading to selective killing of the transformed cells after expression of a prodrug-converting enzyme. Preferential gene expression in tumor versus normal liver tissue was also observed in vivo in a syngeneic rat model. Comparative transduction of a panel of different tumor cell lines with the H1 and the H1/AAV hybrid vector showed a preference of each vector for distinct cell types, probably reflecting the dependence of the viral tropism on capsid determinants.
Collapse
|
8
|
Paglino J, Burnett E, Tattersall P. Exploring the contribution of distal P4 promoter elements to the oncoselectivity of Minute Virus of Mice. Virology 2006; 361:174-84. [PMID: 17175002 PMCID: PMC1853334 DOI: 10.1016/j.virol.2006.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2006] [Revised: 10/13/2006] [Accepted: 11/03/2006] [Indexed: 11/20/2022]
Abstract
Minute Virus of Mice (MVM) shares inherent oncotropic properties with other members of the genus Parvovirus. Two elements responsible, at least in part, for this oncoselectivity have been mapped to an Ets1 binding site adjacent to the P4 TATA box of the initiating promoter, P4, and to a more distal cyclic AMP responsive element (CRE), located within the telomeric hairpin stem. Here the CRE overlaps one half-site for the binding of parvoviral initiation factor (PIF), which is essential for viral DNA replication. We used a degenerate oligonucleotide selection approach to show that CRE binding protein (CREB) selects the sequence ACGTCAC within this context, rather than its more generally accepted palindromic TGACGTCA recognition site. We have developed strategies for manipulating these sequences directly within the left-end palindrome of the MVM infectious clone and used them to clone mutants whose CRE either matches the symmetric consensus sequence or is scrambled, or in which the PIF binding site is incrementally weakened with respect to the CRE. The panel of mutants were tested for fitness relative to wildtype in normal murine fibroblasts A9 or transformed human fibroblasts 324 K, through multiple rounds of growth in co-infected cultures, using a differential real-time quantitative PCR assay. We confirmed that inactivating the CRE substantially abrogates oncoselectivity, but found that improving its fit to the palindromic consensus is somewhat debilitating in either cell type. We also confirmed that reducing the PIF half-site spacing by one basepair enhances oncoselectivity, but found that a further basepair deletion significantly reduces this effect.
Collapse
Affiliation(s)
- Justin Paglino
- Department of Laboratory Medicine, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA
- Graduate Program in Investigative Medicine, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA
| | - Erik Burnett
- Department of Laboratory Genetics, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA
| | - Peter Tattersall
- Department of Laboratory Medicine, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA
- Department of Laboratory Genetics, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA
- * Corresponding author. Department of Laboratory Medicine, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA. Fax: +1 203 688 7340. E-mail address: (P. Tattersall)
| |
Collapse
|
9
|
Li J, Werner E, Hergenhahn M, Poirey R, Luo Z, Rommelaere J, Jauniaux JC. Expression profiling of human hepatoma cells reveals global repression of genes involved in cell proliferation, growth, and apoptosis upon infection with parvovirus H-1. J Virol 2005; 79:2274-86. [PMID: 15681429 PMCID: PMC546555 DOI: 10.1128/jvi.79.4.2274-2286.2005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Autonomous parvoviruses are characterized by their stringent dependency on host cell S phase and their cytopathic effects on neoplastic cells. To better understand the interactions between the virus and its host cell, we used oligonucleotide arrays that carry more than 19,000 unique human gene sequences to profile the gene expression of the human hepatocellular carcinoma cell line QGY-7703 at two time points after parvovirus H-1 infection. At the 6-h time point, a single gene was differentially expressed with a >2.5-fold change. At 12 h, 105 distinct genes were differentially expressed in virus-infected cells compared to mock-treated cells, with 93% of these genes being down-regulated. These repressed genes clustered mainly into classes involved in transcriptional regulation, signal transduction, immune and stress response, and apoptosis, as exemplified by genes encoding the transcription factors Myc, Jun, Fos, Ids, and CEBPs. Quantitative real-time reverse transcription-PCR analysis on selected genes validated the array data and allowed the changes in cellular gene expression to be correlated with the accumulation of viral transcripts and NS1 protein. Western blot analysis of several cellular proteins supported the array results and substantiated the evidence given by these and other data to suggest that the H-1 virus kills QGY-7703 cells by a nonapoptotic process. The promoter regions of most of the differentially expressed genes analyzed fail to harbor any motif for sequence-specific binding of NS1, suggesting that direct binding of NS1 to cellular promoters may not participate in the modulation of cellular gene expression in H-1 virus-infected cells.
Collapse
Affiliation(s)
- Jianhong Li
- Department of Physiology and Biophysics, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
10
|
Davis C, Segev-Amzaleg N, Rotem I, Mincberg M, Amir N, Sivan S, Gitelman I, Tal J. The P4 promoter of the parvovirus minute virus of mice is developmentally regulated in transgenic P4-LacZ mice. Virology 2003; 306:268-79. [PMID: 12642100 DOI: 10.1016/s0042-6822(02)00020-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Activation of the minute virus of mice (MVM) P4 promoter is a key step in the life cycle of the virus and is completely dependent on host transcription factors. Since transcription-factor composition varies widely in different cell types, there is the possibility that only some cell types in the host organism have the capacity to initiate expression from the P4 promoter and therefore that the promoter may be a factor in determining the tropism of MVM. In this study, the ability of various cell types to activate P4, independent of the other virus-host interactions, was examined in transgenic mouse lines bearing a beta-galactosidase reporter sequence driven by the P4 promoter. It was found that lacZ was expressed during embryogenesis and in the adult in a cell-type-specific and differentiation-dependent pattern. The data are consistent with cell-type and stage-specific activation of the P4 promoter having a role in determining the host cell-type range of MVM. The ability of some parvoviruses to replicate in, and kill oncogenically transformed cells, and to destroy induced tumors in laboratory animals is the basis of recent approaches to use MVM-based vectors in cancer gene therapy. Since these vectors rely on the activation of the P4 promoter by the target tissues, understanding the promoter dependence on cell-type and differentiation status is important for their design and potential use.
Collapse
Affiliation(s)
- Claytus Davis
- Department of Molecular Genetics of Development, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Parvoviruses are small, icosahedral viruses (approximately 25 nm) containing a single-strand DNA genome (approximately 5 kb) with hairpin termini. Autonomous parvoviruses (APVs) are found in many species; they do not require a helper virus for replication but they do require proliferating cells (S-phase functions) and, in some cases, tissue-specific factors. APVs can protect animals from spontaneous or experimental tumors, leading to consideration of these viruses, and vectors derived from them, as anticancer agents. Vector development has focused on three rodent APVs that can infect human cells, namely, LuIII, MVM, and H1. LuIII-based vectors with complete replacement of the viral coding sequences can direct transient or persistent expression of transgenes in cell culture. MVM-based and H1-based vectors with substitution of transgenes for the viral capsid sequences retain viral nonstructural (NS) coding sequences and express the NS1 protein. The latter serves to amplify the vector genome in target cells, potentially contributing to antitumor activity. APV vectors have packaging capacity for foreign DNA of approximately 4.8 kb, a limit that probably cannot be exceeded by more than a few percent. LuIII vectors can be pseudotyped with capsid proteins from related APVs, a promising strategy for controlling tissue tropism and circumventing immune responses to repeated administration. Initial success has been achieved in targeting such a pseudotyped vector by genetic modification of the capsid. Subject to advances in production and purification methods, APV vectors have potential as gene transfer agents for experimental and therapeutic use, particularly for cancer therapy.
Collapse
Affiliation(s)
- Ian H Maxwell
- Department of Dermatology and University of Colorado Cancer Center, University of Colorado Health Sciences Center, 4200 East Ninth Avenue, Denver, CO 80262, USA.
| | | | | |
Collapse
|
12
|
Affiliation(s)
- E Sadowy
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | | |
Collapse
|
13
|
Abstract
The concept of using viruses as oncolytic agents has a long history. However, relatively new developments are the use of these viruses as gene delivery vehicles and the restriction of viral replication and lysis to tumour cells. The latter is attempted by the use of tumour-specific promoters, which transcriptionally target viral genes involved in replication, or by deletion of viral functions dispensable for replication in tumour cells but essential for productive infection of normal cells. In addition, retargeting of the viral tropism towards tumours by capsid modifications has been examined. Although much progress has been made in developing oncolytic vectors for clinical use, there is still a long way to go to determine which combinations of virus, gene therapy, surgery, radiation, and/or chemotherapy will provide improved therapy for the control and eradication of a variety of human cancers. First controlled clinical trials with an oncolytic adenovirus in combination with chemotherapy have shown encouraging antineoplastic activity. For future vector developments it will be crucial to achieve maximum vector distribution and transgene expression within tumours, to trigger a specific systemic immune effector response against treated and untreated lesions, and to modulate the immune system to avoid immune-mediated inactivation or destruction of the virus. In the context of replication-competent vectors, suicide genes might be used as fail-safe mechanism in the case of a runaway infection.
Collapse
Affiliation(s)
- O Wildner
- Humboldt-Universität zu Berlin, Labor für Gentherapie, Germany.
| |
Collapse
|
14
|
Clément N, Avalosse B, El Bakkouri K, Velu T, Brandenburger A. Cloning and sequencing of defective particles derived from the autonomous parvovirus minute virus of mice for the construction of vectors with minimal cis-acting sequences. J Virol 2001; 75:1284-93. [PMID: 11152501 PMCID: PMC114034 DOI: 10.1128/jvi.75.3.1284-1293.2001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The production of wild-type-free stocks of recombinant parvovirus minute virus of mice [MVM(p)] is difficult due to the presence of homologous sequences in vector and helper genomes that cannot easily be eliminated from the overlapping coding sequences. We have therefore cloned and sequenced spontaneously occurring defective particles of MVM(p) with very small genomes to identify the minimal cis-acting sequences required for DNA amplification and virus production. One of them has lost all capsid-coding sequences but is still able to replicate in permissive cells when nonstructural proteins are provided in trans by a helper plasmid. Vectors derived from this particle produce stocks with no detectable wild-type MVM after cotransfection with new, matched, helper plasmids that present no homology downstream from the transgene.
Collapse
Affiliation(s)
- N Clément
- IRIBHN-IBMM, Université Libre de Bruxelles, B-6041 Gosselies, Belgium
| | | | | | | | | |
Collapse
|
15
|
Dupont F, Avalosse B, Karim A, Mine N, Bosseler M, Maron A, Van den Broeke AV, Ghanem GE, Burny A, Zeicher M. Tumor-selective gene transduction and cell killing with an oncotropic autonomous parvovirus-based vector. Gene Ther 2000; 7:790-6. [PMID: 10822306 DOI: 10.1038/sj.gt.3301161] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A recombinant MVMp of the fibrotropic strain of minute virus of mice (MVMp) expressing the chloramphenicol acetyltransferase reporter gene was used to infect a series of biologically relevant cultured cells, normal or tumor-derived, including normal melanocytes versus melanoma cells, normal mammary epithelial cells versus breast adenocarcinoma cells, and normal neurons or astrocytes versus glioma cells. As a reference cell system we used normal human fibroblasts versus the SV40-transformed fibroblast cell line NB324K. After infection, we observed good expression of the reporter gene in the different tumor cell types, but only poor expression if any in the corresponding normal cells. We also constructed a recombinant MVMp expressing the green fluorescent protein reporter gene and assessed by flow cytometry the efficiency of gene transduction into the different target cells. At a multiplicity of infection of 30, we observed substantial transduction of the gene into most of the tumor cell types tested, but only marginal transduction into normal cells under the same experimental conditions. Finally, we demonstrated that a recombinant MVMp expressing the herpes simplex virus thymidine kinase gene can, in vitro, cause efficient killing of most tumor cell types in the presence of ganciclovir, whilst affecting normal proliferating cells only marginally if at all. However, in the same experimental condition, breast tumor cells appeared to be resistant to GCV-mediated cytotoxicity, possibly because these cells are not susceptible to the bystander effect. Our data suggest that MVMp-based vectors could prove useful as selective vehicles for anticancer gene therapy, particularly for in vivo delivery of cytotoxic effector genes into tumor cells.
Collapse
Affiliation(s)
- F Dupont
- Laboratoire d'Investigation Clinique et d'Oncologie Expérimentale, Unité d'Oncologie Moléculaire, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Perros M, Fuks F, Kherrouche Z, Rommelaere J. Atypical nucleoprotein complexes mediate CRE-dependent regulation of the early promoter of minute virus of mice. J Gen Virol 1999; 80 ( Pt 12):3267-3272. [PMID: 10567660 DOI: 10.1099/0022-1317-80-12-3267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The P4 promoter of the parvovirus minute virus of mice (MVMp) directs transcription of the genes encoding non-structural proteins. We have previously shown that functional upstream CRE elements contribute to both the ras oncogene-dependent activation of promoter P4 and its down-modulation by known activators of cyclic AMP-dependent protein kinase A (PKA). In the present work, the nucleoprotein complexes formed with the P4 CRE elements were characterized with regard to their polypeptide constituents and the nucleotides taking part in the interaction. Atypical interactions, both at the protein-protein and protein-DNA level, were observed, which may be a reflection of the divergence of the parvoviral CREs from the usual consensus. The CRE-mediated regulation of promoter P4 by PKA and Ras is discussed in light of these findings.
Collapse
Affiliation(s)
- Manoussos Perros
- Applied Tumour Virology, Abteilung F0100 and Institut National de la Santé et de la Recherche Médicale U375, Deutsches Krebsforschungszentrum, Postfach 10 19 49, D-69009 Heidelberg, Germany1
| | - François Fuks
- Applied Tumour Virology, Abteilung F0100 and Institut National de la Santé et de la Recherche Médicale U375, Deutsches Krebsforschungszentrum, Postfach 10 19 49, D-69009 Heidelberg, Germany1
| | - Zoulika Kherrouche
- Molecular Oncology Unit, UMR 8526, Institut Pasteur de Lille, F-59019 Lille, France2
| | - Jean Rommelaere
- Applied Tumour Virology, Abteilung F0100 and Institut National de la Santé et de la Recherche Médicale U375, Deutsches Krebsforschungszentrum, Postfach 10 19 49, D-69009 Heidelberg, Germany1
| |
Collapse
|
17
|
Deleu L, Pujol A, Faisst S, Rommelaere J. Activation of promoter P4 of the autonomous parvovirus minute virus of mice at early S phase is required for productive infection. J Virol 1999; 73:3877-85. [PMID: 10196282 PMCID: PMC104165 DOI: 10.1128/jvi.73.5.3877-3885.1999] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Autonomous parvoviruses are tightly dependent on host cell factors for various steps of their life cycle. In particular, DNA replication and gene expression of the prototype strain of the minute virus of mice (MVMp) are closely linked to the onset of host cell DNA replication, pointing to the involvement of an S-phase-specific cellular factor(s) in parvovirus multiplication. The viral nonstructural protein NS-1 is absolutely required for parvovirus DNA replication and is able to transcriptionally regulate parvoviral and heterologous promoters. We previously showed that the promoter P4, which directs the transcription unit encoding the NS proteins, is activated at the onset of S phase. This activation is dependent on an E2F motif in the proximal region of promoter P4. An infectious MVM DNA clone was mutated in the E2F motif of P4. The wild type and the E2F mutant derivative were tested for their ability to produce progeny viruses after transfection of permissive cells. In the context of the whole MVMp genome, the E2F mutation abolished P4 induction in S phase and inactivated the infectious molecular clone, which failed to become amplified and generate progeny particles. The virus could be rescued when NS proteins were supplied in trans, showing that P4 hyperactivity in S is needed to reach a level of NS-1 expression that is sufficient to drive the viral replication cycle. These data show that E2F-mediated P4 activation at the early S phase is a limiting factor for parvovirus production. The primary barrier to parvovirus gene expression in G1 is thought to be promoter formation rather than activation, due to the poor conversion of the parental single-strand genome to a duplex form. The S dependence of P4 activation may therefore be a sign of the virus adaptation to life in the S-phase host cell. If the conversion block in G1 were to be leaky, the S induction of promoter P4 could be envisioned as a safeguard against the production of toxic NS proteins until cells reach the S phase and provide the full machinery for parvovirus replication.
Collapse
Affiliation(s)
- L Deleu
- Applied Tumor Virology, Abteilung F0100 and Institut National de la Santé et de la Recherche Médicale U375, Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
18
|
Karantzoulis-Fegaras F, Antoniou H, Lai SL, Kulkarni G, D'Abreo C, Wong GK, Miller TL, Chan Y, Atkins J, Wang Y, Marsden PA. Characterization of the human endothelial nitric-oxide synthase promoter. J Biol Chem 1999; 274:3076-93. [PMID: 9915847 DOI: 10.1074/jbc.274.5.3076] [Citation(s) in RCA: 160] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Understanding transcription initiation of the endothelial nitric-oxide synthase (eNOS) gene appears pivotal to gaining a comprehensive view of NO biology in the blood vessel wall. The present study therefore focused upon a detailed dissection of the functionally important cis-DNA elements and the multiprotein complexes implicated in the cooperative control of constitutive expression of the human eNOS gene in vascular endothelium. Two tightly clustered cis-regulatory regions were identified in the proximal enhancer of the TATA-less eNOS promoter using deletion analysis and linker-scanning mutagenesis: positive regulatory domains I (-104/-95 relative to transcription initiation) and II (-144/-115). Analysis of trans-factor binding and functional expression studies revealed a surprising degree of cooperativity and complexity. The nucleoprotein complexes that form upon these regions in endothelial cells contained Ets family members, Sp1, variants of Sp3, MAZ, and YY1. Functional domain studies in Drosophila Schneider cells and endothelial cells revealed examples of positive and negative protein-protein cooperativity involving Sp1, variants of Sp3, Ets-1, Elf-1, and MAZ. Therefore, multiprotein complexes are formed on the activator recognition sites within this 50-base pair region of the human eNOS promoter in vascular endothelium.
Collapse
Affiliation(s)
- F Karantzoulis-Fegaras
- Renal Division and Department of Medicine, St. Michael's Hospital and University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Vassias I, Hazan U, Michel Y, Sawa C, Handa H, Gouya L, Morinet F. Regulation of human B19 parvovirus promoter expression by hGABP (E4TF1) transcription factor. J Biol Chem 1998; 273:8287-93. [PMID: 9525935 DOI: 10.1074/jbc.273.14.8287] [Citation(s) in RCA: 21] [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
The genetic expression of human B19 parvovirus is only dependent on one promoter in vivo and in vitro. This is the P6 promoter, which is located on the left side of the genome and is a single-stranded DNA molecule. This led us to investigate the regulation of the P6 promoter and the possible resulting variability of the nucleotide sequence. After analysis of the promoter region of 17 B19 strains, only 1.5% variability was found. More exciting was the finding of mutations that were clustered around the TATA box and defined a highly conserved region (nucleotides 113-210) in the proximal part of the P6 promoter. HeLa and UT7/Epo cell extracts were found to protect this region, which contained a core motif for Ets family proteins, with YY1 and Sp1 binding sites on either side. Gel mobility shift assays performed with nuclear proteins from HeLa and UT7/Epo cells identified DNA-binding proteins specific for these sites. By supershift analysis, we demonstrated the binding of the hGABP (also named E4TF1) protein to the Ets binding site and the fixation of Sp1 and YY1 proteins on their respective motifs. In Drosophila SL2 cells, hGABPalpha and -beta stimulated P6 promoter activity, and hGABPalpha/hGABPbeta and Sp1 exerted synergistic stimulation of this activity, an effect diminished by YY1.
Collapse
Affiliation(s)
- I Vassias
- Service de Microbiologie, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75475 Paris CEDEX 10, France
| | | | | | | | | | | | | |
Collapse
|
20
|
Faisst S, Guittard D, Benner A, Cesbron JY, Schlehofer JR, Rommelaere J, Dupressoir T. Dose-dependent regression of HeLa cell-derived tumours in SCID mice after parvovirus H-1 infection. Int J Cancer 1998; 75:584-9. [PMID: 9466660 DOI: 10.1002/(sici)1097-0215(19980209)75:4<584::aid-ijc15>3.0.co;2-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Parvoviruses of rodents are endowed with oncosuppressive properties. In particular, parvoviral infections protect host animals from spontaneous and chemical- or virus-induced tumour initiation in laboratory animals. The present study was undertaken to substantiate the capacity of parvovirus H-1 to inhibit therapeutically the growth of established tumours originating from human carcinoma cells implanted in recipient mice. To this end, quickly growing s.c. carcinomas were established by injection of human cervical carcinoma cells (HeLa) into immunodeficient (SCID) mice. Tumour-bearing mice subsequently were inoculated with H-1 at various multiplicities of infection. H-1 virus infection led to regression of tumours, the onset and efficiency of which were dose-dependent.
Collapse
Affiliation(s)
- S Faisst
- Tumour Virology Unit and INSERM U 375, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
21
|
Lorson C, Pearson J, Burger L, Pintel DJ. An Sp1-binding site and TATA element are sufficient to support full transactivation by proximally bound NS1 protein of minute virus of mice. Virology 1998; 240:326-37. [PMID: 9454706 DOI: 10.1006/viro.1997.8940] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The minute virus of mice (MVM) P38 Sp1-binding site and TATA box, inserted in an otherwise heterologous plasmid background, could be transactivated to high levels by the MVM NS1 protein targeted proximally to these sequences, demonstrating that these core promoter regulatory elements are sufficient to support essentially wild-type levels of NS1-transactivated expression and suggesting that NS1 may act directly or indirectly with Sp1 and or elements of the general transcription machinery. Accordingly, we show that bacterially generated NS1 can interact strongly, independent of nucleic acid bridging, and most likely directly with Sp1 in vitro and can associate, in a nucleic acid-independent manner, with endogenous Sp1 as it exists in a complex transcriptionally active murine nuclear extract NS1 achieves the same fold activation of an isolated TATA element over its low basal level and can also be demonstrated to interact efficiently and specifically with the general transcription factors TBP and TFIIA (alpha, beta) in vitro.
Collapse
Affiliation(s)
- C Lorson
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri-Columbia 65212, USA
| | | | | | | |
Collapse
|
22
|
Deleu L, Fuks F, Spitkovsky D, Hörlein R, Faisst S, Rommelaere J. Opposite transcriptional effects of cyclic AMP-responsive elements in confluent or p27KIP-overexpressing cells versus serum-starved or growing cells. Mol Cell Biol 1998; 18:409-19. [PMID: 9418888 PMCID: PMC121511 DOI: 10.1128/mcb.18.1.409] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The minute virus of mice, an autonomous parvovirus, requires entry of host cells into the S phase of the cell cycle for its DNA to be amplified and its genes expressed. This work focuses on the P4 promoter of this parvovirus, which directs expression of the transcription unit encoding the parvoviral nonstructural polypeptides. These notably include protein NS1, necessary for the S-phase-dependent burst of parvoviral DNA amplification and gene expression. The activity of the P4 promoter is shown to be regulated in a cell cycle-dependent manner. At the G1/S-phase transition, the promoter is activated via a cis-acting DNA element which interacts with phase-specific complexes containing the cellular transcription factor E2F. It is inhibited, on the other hand, in cells arrested in G1 due to contact inhibition. This inhibitory effect is not observed in serum-starved cells. It is mediated in cis by cyclic AMP response elements (CREs). Unlike serum-starved cells, confluent cells accumulate the cyclin-dependent kinase inhibitor p27, suggesting that the switch from CRE-mediated activation to CRE-mediated repression involves the p27 protein. Accordingly, plasmid-driven overexpression of p27 causes down-modulation of promoter P4 in growing cells, depending on the presence of at least two functional CREs. No such effect is observed with two other cyclin-dependent kinase inhibitors, p16 and p21. Given the importance of P4-driven synthesis of protein NS1 in parvoviral DNA amplification and gene expression, the stringent S-phase dependency of promoter P4 is likely a major determinant of the absolute requirement of the minute virus of mice for host cell proliferation.
Collapse
Affiliation(s)
- L Deleu
- Applied Tumor Virology, Abteilung 0610 and Institut National de la Santé et de la Recherche Médicale U 375, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
23
|
Karperien M, Farih-Sips H, Löwik CW, de Laat SW, Boonstra J, Defize LH. Expression of the parathyroid hormone-related peptide gene in retinoic acid-induced differentiation: involvement of ETS and Sp1. Mol Endocrinol 1997; 11:1435-48. [PMID: 9280059 DOI: 10.1210/mend.11.10.9997] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Differentiation of P19 embryonal carcinoma (EC) and embryonal stem (ES)-5 cells with retinoic acid (RA) induces expression of PTH-related peptide (PTHrP) mRNA. In this study we have characterized a region between nucleotide (nt) -88 and -58 relative to the transcription start site in the murine PTHrP gene that was involved in this expression. Sequence analysis identified two partially overlapping binding sites for the Ets family of transcription factors and an inverted Sp1-binding site. Two major specific bands were detected in a bandshift assay using an oligonucleotide spanning nt -88 and -58 as a probe and nuclear extracts from both undifferentiated and RA-differentiated P19 EC cells. The lower complex consisted of Ets-binding proteins as demonstrated by competition with consensus Ets-binding sites, while the upper complex contained Sp1-binding activity as demonstrated by competition with consensus Sp1-binding sites. The observed bandshift patterns using nuclear extracts of undifferentiated or RA-differentiated P19 cells were indistinguishable, suggesting that the differentiation-mediated expression was not caused by the induction of expression of new transcription factors. Mutations in either of the Ets-binding sites or the Sp1-binding site completely abolished RA-induced expression of PTHrP promoter reporter constructs, indicating that the RA effect was dependent on the simultaneous action of both Ets- and Sp1-like activities. Furthermore, these mutations also abolished promoter activity in cells that constitutively expressed PTHrP mRNA, suggesting a central role for the Ets and Sp1 families of transcription factors in the expression regulation of the mouse PTHrP gene.
Collapse
Affiliation(s)
- M Karperien
- Department of Endocrinology, Leiden University, The Netherlands
| | | | | | | | | | | |
Collapse
|
24
|
Abstract
While the minute virus of mice (MVM) P4 promoter, which drives the viral nonstructural genes, is highly active in the absence of viral proteins, P38, the capsid gene promoter, is strictly dependent on the viral nonstructural protein NS1. Once fully transactivated, however, P38 mediates twice the steady-state level of expression achieved by P4. In this report, we address the discrepancy between the ability of P38 to mediate very high levels of activated transcription yet only low levels of basal expression, and we investigate the determinants that govern P38 basal expression. The isolated P38 core promoter elements (the P38 Sp1-binding site and TATA element) are at least as transcriptionally competent as the analogous P4 promoter elements. Proximally positioning P4 enhancer factor-binding sequences (nucleotides [nt] 57 to 157) upstream of isolated P38 core transcription regulatory elements or upstream of a native, though abbreviated, P38 cassette (MVM nt 1938 to 2072) confers significant levels of expression to P38 in the absence of NS1, while the full left-end hairpin sequences (nt 1 to 133) elevate basal P38 activity to levels equivalent to P4 basal levels. In the context of the complete viral genome, however, proximally positioned enhancer sequences are unable to confer significant levels of expression to P38, suggesting that low P38 basal levels are a consequence not only of a lack of proximal enhancer elements but also of additional positional regulatory constraints which can be overcome by NS1.
Collapse
Affiliation(s)
- C Lorson
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia 65212, USA
| | | |
Collapse
|
25
|
Anouja F, Wattiez R, Mousset S, Caillet-Fauquet P. The cytotoxicity of the parvovirus minute virus of mice nonstructural protein NS1 is related to changes in the synthesis and phosphorylation of cell proteins. J Virol 1997; 71:4671-8. [PMID: 9151861 PMCID: PMC191689 DOI: 10.1128/jvi.71.6.4671-4678.1997] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Autonomous parvoviruses exert lytic and cytostatic effects believed to contribute to their antineoplastic activity. Studies with inducible clones have demonstrated a direct involvement of parvovirus nonstructural proteins (NS) in oncolysis. Human and rat fibroblasts have been stably transfected with MVM(p) (minute virus of mice prototype strain) NS genes cloned under the control of a hormone-inducible promoter. Dexamethasone-induced synthesis of the NS proteins in sensitive transformed cells results in cell killing within a few days. From these sensitive cell lines have been isolated some NS-resistant clones that also prove resistant to MVM(p) infection, suggesting that cell factors modulate NS cytotoxicity. We have previously reported that factors involved in cell cycle regulation may contribute to this modulation, since NS toxicity requires cell proliferation and correlates with a cell cycle perturbation leading to an arrest in phase S/G2. In addition to its role in cytotoxicity, NS1 can regulate transcription driven by parvovirus and nonparvovirus promoters. Since phosphorylation is a critical event in controlling the activity of many proteins, notably transcription factors and cell cycle-regulated proteins, we have examined the effect of NS1 on the synthesis and phosphorylation of cell proteins. Our results indicate that NS1 interferes, within 7 h of induction, with phosphorylation of a protein of about 14 kDa (p14). Cell synchronization has enabled us to show that phosphorylation of this protein occurs in early S phase and is prevented when NS1 is induced. This early effect of NS1 on p14 phosphorylation may be directly linked to cytotoxicity and is probably related to the previously reported inhibition of cell DNA synthesis. Late in the induction period (24 h), NS1 also alters the synthesis of a 50-kDa protein and a 35-kDa protein (p50 and p35, respectively). Microsequencing of p35 reveals sequence homology with beta-tubulin. These effects of NS1, observed only in NS1-sensitive cell lines, may be related to the protein's cytotoxicity.
Collapse
Affiliation(s)
- F Anouja
- Département de Biologie Moléculaire, Université Libre de Bruxelles, Rhode-St-Genèse, Belgium
| | | | | | | |
Collapse
|
26
|
Dittmer J, Pise-Masison CA, Clemens KE, Choi KS, Brady JN. Interaction of human T-cell lymphotropic virus type I Tax, Ets1, and Sp1 in transactivation of the PTHrP P2 promoter. J Biol Chem 1997; 272:4953-8. [PMID: 9030555 DOI: 10.1074/jbc.272.8.4953] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We have previously shown that the parathyroid hormone-related protein (PTHrP) promoter contains binding sites for transcription factors Ets1 and Sp1 and that human T-cell lymphotropic virus type I (HTLV-I) Tax cooperates with Ets1 to transactivate the PTHrP P2 promoter. Using the yeast two-hybrid interaction system, we now provide evidence that Tax interacts with Ets1. Moreover, a double mutation (D22A,C23S) in the Tax protein that abrogated the Tax/Ets1 interaction also inhibited the Tax/Ets1 cooperative effect, suggesting that the interaction between Tax and Ets1 is important for transactivation of the PTHrP promoter. In coimmunoprecipitation assays, we find that Tax facilitates the interaction between Ets1 and Sp1, forming a ternary complex. When the Sp1 site in the PTHrP promoter was mutated, the Tax/Ets1 cooperative effect was dramatically decreased. This suggests that Sp1 plays an important role in the Ets1-dependent Tax transactivation of the PTHrP P2 promoter. Finally, we demonstrate that Gal4-Tax is a strong activator of the Gal PTHrP promoter, implying that Tax contributes directly to the transcriptional activation of the promoter. We propose a model in which the Tax/Ets1 cooperative effect on the PTHrP P2 promoter is based on the ability of Tax, Ets1, and Sp1 to form a ternary complex on the template DNA. Tax facilitates the interaction of Ets1/Sp1 and participates directly in the transcription initiation process.
Collapse
Affiliation(s)
- J Dittmer
- Virus Tumor Biology Section, Laboratory of Molecular Virology, NCI, National Institutes of Health, Bethesda, Maryland 20892-5005, USA
| | | | | | | | | |
Collapse
|