Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Virol ; 93(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30842317

ABSTRACT

The adenovirus (Ad) E4orf4 protein contributes to virus-induced inhibition of the DNA damage response (DDR) by reducing ATM and ATR signaling. Consequently, E4orf4 inhibits DNA repair and sensitizes transformed cells to killing by DNA-damaging drugs. Inhibition of ATM and ATR signaling contributes to the efficiency of virus replication and may provide one explanation for the cancer selectivity of cell death induced by the expression of E4orf4 alone. In this report, we investigate a direct interaction of E4orf4 with the DDR. We show that E4orf4 physically associates with the DNA-dependent protein kinase (DNA-PK), and we demonstrate a biphasic functional interaction between these proteins, wherein DNA-PK is required for ATM and ATR inhibition by E4orf4 earlier during infection but is inhibited by E4orf4 as infection progresses. This biphasic process is accompanied by initial augmentation and a later inhibition of DNA-PK autophosphorylation as well as by colocalization of DNA-PK with early Ad replication centers and distancing of DNA-PK from late replication centers. Moreover, inhibition of DNA-PK improves Ad replication more effectively when a DNA-PK inhibitor is added later rather than earlier during infection. When expressed alone, E4orf4 is recruited to DNA damage sites in a DNA-PK-dependent manner. DNA-PK inhibition reduces the ability of E4orf4 to induce cancer cell death, likely because E4orf4 is prevented from arriving at the damage sites and from inhibiting the DDR. Our results support an important role for the E4orf4-DNA-PK interaction in Ad replication and in facilitation of E4orf4-induced cancer-selective cell death.IMPORTANCE Several DNA viruses evolved mechanisms to inhibit the cellular DNA damage response (DDR), which acts as an antiviral defense system. We present a novel mechanism by which the adenovirus (Ad) E4orf4 protein inhibits the DDR. E4orf4 interacts with the DNA damage sensor DNA-PK in a biphasic manner. Early during infection, E4orf4 requires DNA-PK activity to inhibit various branches of the DDR, whereas it later inhibits DNA-PK itself. Furthermore, although both E4orf4 and DNA-PK are recruited to virus replication centers (RCs), DNA-PK is later distanced from late-phase RCs. Delayed DNA-PK inhibition greatly contributes to Ad replication efficiency. When E4orf4 is expressed alone, it is recruited to DNA damage sites. Inhibition of DNA-PK prevents both recruitment and the previously reported ability of E4orf4 to kill cancer cells. Our results support an important role for the E4orf4-DNA-PK interaction in Ad replication and in facilitation of E4orf4-induced cancer-selective cell death.


Subject(s)
DNA Damage/physiology , DNA-Activated Protein Kinase/metabolism , Viral Proteins/metabolism , Adenoviridae/genetics , Adenoviridae Infections/genetics , Adenovirus E4 Proteins/metabolism , Adenovirus E4 Proteins/physiology , Adenoviruses, Human/physiology , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , DNA Repair/physiology , DNA, Viral/genetics , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/physiology , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Nuclear Proteins/metabolism , Phosphorylation , Signal Transduction , Viral Proteins/physiology , Virus Replication/physiology
2.
J Virol ; 93(19)2019 10 01.
Article in English | MEDLINE | ID: mdl-31315986

ABSTRACT

The adenovirus (Ad) E4orf4 protein was reported to contribute to inhibition of ATM- and ATR-regulated DNA damage signaling during Ad infection and following treatment with DNA-damaging drugs. Inhibition of these pathways improved Ad replication, and when expressed alone, E4orf4 sensitized transformed cells to drug-induced toxicity. However, the mechanisms utilized were not identified. Here, we show that E4orf4 associates with the DNA damage sensor poly(ADP-ribose) polymerase 1 (PARP-1) and that the association requires PARP activity. During Ad infection, PARP is activated, but its activity is not required for recruitment of either E4orf4 or PARP-1 to virus replication centers, suggesting that their association occurs following recruitment. Inhibition of PARP-1 assists E4orf4 in reducing DNA damage signaling during infection, and E4orf4 attenuates virus- and DNA damage-induced parylation. Furthermore, E4orf4 reduces PARP-1 phosphorylation on serine residues, which likely contributes to PARP-1 inhibition as phosphorylation of this enzyme was reported to enhance its activity. PARP-1 inhibition is important to Ad infection since treatment with a PARP inhibitor enhances replication efficiency. When E4orf4 is expressed alone, it associates with poly(ADP-ribose) (PAR) chains and is recruited to DNA damage sites in a PARP-1-dependent manner. This recruitment is required for inhibition of drug-induced ATR signaling by E4orf4 and for E4orf4-induced cancer cell death. Thus, the results presented here demonstrate a novel mechanism by which E4orf4 targets and inhibits DNA damage signaling through an association with PARP-1 for the benefit of the virus and impacting E4orf4-induced cancer cell death.IMPORTANCE Replication intermediates and ends of viral DNA genomes can be recognized by the cellular DNA damage response (DDR) network as DNA damage whose repair may lead to inhibition of virus replication. Therefore, many viruses evolved mechanisms to inhibit the DDR network. We have previously shown that the adenovirus (Ad) E4orf4 protein inhibits DDR signaling, but the mechanisms were not identified. Here, we describe an association of E4orf4 with the DNA damage sensor poly(ADP-ribose) polymerase 1 (PARP-1). E4orf4 reduces phosphorylation of this enzyme and inhibits its activity. PARP-1 inhibition assists E4orf4 in reducing Ad-induced DDR signaling and improves the efficiency of virus replication. Furthermore, the ability of E4orf4, when expressed alone, to accumulate at DNA damage sites and to kill cancer cells is attenuated by chemical inhibition of PARP-1. Our results indicate that the E4orf4-PARP-1 interaction has an important role in Ad replication and in promotion of E4orf4-induced cancer-selective cell death.


Subject(s)
Adenoviridae/growth & development , DNA Damage , Host-Pathogen Interactions , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly ADP Ribosylation , Signal Transduction , Viral Proteins/metabolism , Cell Line , Humans , Virus Replication
4.
PLoS Pathog ; 12(2): e1005420, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26867009

ABSTRACT

The DNA damage response (DDR) is a conglomerate of pathways designed to detect DNA damage and signal its presence to cell cycle checkpoints and to the repair machinery, allowing the cell to pause and mend the damage, or if the damage is too severe, to trigger apoptosis or senescence. Various DDR branches are regulated by kinases of the phosphatidylinositol 3-kinase-like protein kinase family, including ataxia-telangiectasia mutated (ATM) and ATM- and Rad3-related (ATR). Replication intermediates and linear double-stranded genomes of DNA viruses are perceived by the cell as DNA damage and activate the DDR. If allowed to operate, the DDR will stimulate ligation of viral genomes and will inhibit virus replication. To prevent this outcome, many DNA viruses evolved ways to limit the DDR. As part of its attack on the DDR, adenovirus utilizes various viral proteins to cause degradation of DDR proteins and to sequester the MRN damage sensor outside virus replication centers. Here we show that adenovirus evolved yet another novel mechanism to inhibit the DDR. The E4orf4 protein, together with its cellular partner PP2A, reduces phosphorylation of ATM and ATR substrates in virus-infected cells and in cells treated with DNA damaging drugs, and causes accumulation of damaged DNA in the drug-treated cells. ATM and ATR are not mutually required for inhibition of their signaling pathways by E4orf4. ATM and ATR deficiency as well as E4orf4 expression enhance infection efficiency. Furthermore, E4orf4, previously reported to induce cancer-specific cell death when expressed alone, sensitizes cells to killing by sub-lethal concentrations of DNA damaging drugs, likely because it inhibits DNA damage repair. These findings provide one explanation for the cancer-specificity of E4orf4-induced cell death as many cancers have DDR deficiencies leading to increased reliance on the remaining intact DDR pathways and to enhanced susceptibility to DDR inhibitors such as E4orf4. Thus DDR inhibition by E4orf4 contributes both to the efficiency of adenovirus replication and to the ability of E4orf4 to kill cancer cells.


Subject(s)
Adenoviridae Infections/virology , Adenoviruses, Human/physiology , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Damage , Viral Proteins/metabolism , Adenoviruses, Human/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Death , Cell Line, Tumor , DNA Repair , DNA Replication , Humans , Mutation , Phosphorylation , Signal Transduction , Viral Proteins/genetics , Virus Replication
5.
Proc Natl Acad Sci U S A ; 110(19): E1724-33, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23613593

ABSTRACT

The adenovirus E4orf4 protein regulates the progression of viral infection, and when expressed alone in mammalian tissue culture cells it induces protein phosphatase 2A (PP2A)-B55- and Src-dependent cell death, which is more efficient in oncogene-transformed cells than in normal cells. This form of cell death is caspase-independent, although it interacts with classic caspase-dependent apoptosis. PP2A-B55-dependent E4orf4-induced toxicity is highly conserved in evolution from yeast to mammalian cells. In this work we investigated E4orf4-induced cell death in a whole multicellular organism, Drosophila melanogaster. We show that E4orf4 induced low levels of cell killing, caused by both caspase-dependent and -independent mechanisms. Drosophila PP2A-B55 (twins/abnormal anaphase resolution) and Src64B contributed additively to this form of cell death. Our results provide insight into E4orf4-induced cell death, demonstrating that in parallel to activating caspase-dependent apoptosis, E4orf4 also inhibited this form of cell death induced by the proapoptotic genes reaper, head involution defective, and grim. The combination of both induction and inhibition of caspase-dependent cell death resulted in low levels of tissue damage that may explain the inefficient cell killing induced by E4orf4 in normal cells in tissue culture. Furthermore, E4orf4 inhibited JNK-dependent cell killing as well. However, JNK inhibition did not impede E4orf4-induced toxicity and even enhanced it, indicating that E4orf4-induced cell killing is a distinctive form of cell death that differs from both JNK- and Rpr/Hid/Grim-induced forms of cell death.


Subject(s)
Apoptosis , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation, Enzymologic , Phosphoprotein Phosphatases/metabolism , Viral Proteins/genetics , src-Family Kinases/metabolism , Animals , Caspases/metabolism , Conserved Sequence , Female , Male , Models, Genetic , Phenotype , Photoreceptor Cells, Invertebrate/physiology , Transgenes , Viral Proteins/metabolism , Wings, Animal/physiology
6.
J Virol ; 88(11): 6318-28, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24672025

ABSTRACT

UNLABELLED: The adenovirus E4orf4 protein induces nonclassical apoptosis in mammalian cells through at least two complementing pathways regulated by the interactions of E4orf4 with protein phosphatase 2A (PP2A) and Src kinases. In Saccharomyces cerevisiae cells, which do not express Src, E4orf4 induces PP2A-dependent toxicity. The yeast Golgi apyrase Ynd1 was found to contribute to E4orf4-mediated toxicity and to interact with the PP2A-B55α regulatory subunit. In addition, a mammalian Ynd1 orthologue, the NTPDASE4 gene product Golgi UDPase, was shown to physically interact with E4orf4. Here we report that knockdown of NTPDASE4 suppressed E4orf4-induced cell death. Conversely, overexpression of the NTPDASE4 gene products Golgi UDPase and LALP70 enhanced E4orf4-induced cell killing. We found that similarly to results obtained in yeast, the apyrase activity of mammalian UDPase was not required for its contribution to E4orf4-induced toxicity. The interaction between E4orf4 and UDPase had two consequences: a PP2A-dependent one, resulting in increased UDPase levels, and a PP2A-independent outcome that led to dissociation of large UDPase-containing protein complexes. The present report extends our findings in yeast to E4orf4-mediated death of mammalian cells, and combined with previous results, it suggests that the E4orf4-NTPDase4 pathway, partly in association with PP2A, may provide an alternative mechanism for the E4orf4-Src pathway to contribute to the cytoplasmic death function of E4orf4. IMPORTANCE: The adenovirus E4orf4 protein contributes to regulation of the progression of virus infection from the early to the late phase, and when expressed alone, it induces a unique caspase-independent programmed cell death which is more efficient in cancer cells than in normal cells. The interactions of E4orf4 with cellular proteins that mediate its functions, such as PP2A and Src kinases, are highly conserved in evolution. The results presented here reveal that the NTPDASE4 gene product Golgi UDPase, first discovered to contribute to E4orf4 toxicity in Saccharomyces cerevisiae, associates with E4orf4 and plays a role in induction of cell death in mammalian cells. Details of the functional interaction between E4orf4, PP2A, and the UDPase are described. Identification of the evolutionarily conserved mechanisms underlying E4orf4 activity will increase our understanding of the interactions between the virus and the host cell and will contribute to our grasp of the unique mode of E4orf4-induced cell death.


Subject(s)
Apoptosis/genetics , Apyrase/metabolism , Nucleoside-Triphosphatase/metabolism , Protein Phosphatase 2/metabolism , Pyrophosphatases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Viral Proteins/metabolism , Apoptosis/physiology , Blotting, Western , Chromatography, Gel , DNA Primers/genetics , Gene Knockdown Techniques , Glutathione Transferase , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Image Processing, Computer-Assisted , Immunoprecipitation , Plasmids/genetics , Pyrophosphatases/genetics , Saccharomyces cerevisiae , Viral Proteins/toxicity
7.
J Biol Chem ; 288(19): 13718-27, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23530045

ABSTRACT

BACKGROUND: The adenovirus E4orf4 protein must bind protein phosphatase 2A (PP2A) for its functions. RESULTS: The E4orf4 binding site in PP2A was mapped to the α1,α2 helices of the B55α subunit. CONCLUSION: The E4orf4 binding site in PP2A-B55α lies above the substrate binding site and does not overlap it. SIGNIFICANCE: A novel functional significance was assigned to the α1,α2 helices of the PP2A-B55α subunit. The adenovirus E4orf4 protein regulates the progression of viral infection and when expressed outside the context of the virus it induces nonclassical, cancer cell-specific apoptosis. All E4orf4 functions known to date require an interaction between E4orf4 and protein phosphatase 2A (PP2A), which is mediated through PP2A regulatory B subunits. Specifically, an interaction with the B55α subunit is required for induction of cell death by E4orf4. To gain a better insight into the E4orf4-PP2A interaction, mapping of the E4orf4 interaction site in PP2A-B55α has been undertaken. To this end we used a combination of bioinformatics analyses of PP2A-B55α and of E4orf4, which led to the prediction of E4orf4 binding sites on the surface of PP2A-B55α. Mutation analysis, immunoprecipitation, and GST pulldown assays based on the theoretical predictions revealed that the E4orf4 binding site included the α1 and α2 helices described in the B55α structure and involved at least three residues located in these helices facing each other. Loss of E4orf4 binding was accompanied by reduced contribution of the B55α mutants to E4orf4-induced cell death. The identified E4orf4 binding domain lies above the previously described substrate binding site and does not overlap it, although its location could be consistent with direct or indirect effects on substrate binding. This work assigns for the first time a functional significance to the α1,α2 helices of B55α, and we suggest that the binding site defined by these helices could also contribute to interactions between PP2A and some of its cellular regulators.


Subject(s)
Adenovirus E4 Proteins/chemistry , Protein Phosphatase 2/chemistry , Amino Acid Motifs , Amino Acid Substitution , Animals , Apoptosis , Binding Sites , Conserved Sequence , HEK293 Cells , Humans , Molecular Docking Simulation , Protein Binding , Protein Interaction Domains and Motifs , Protein Phosphatase 2/genetics , Protein Structure, Secondary , Rats , Structural Homology, Protein
8.
Adv Exp Med Biol ; 818: 61-97, 2014.
Article in English | MEDLINE | ID: mdl-25001532

ABSTRACT

The adenovirus E4orf4 protein is a multifunctional viral regulator that contributes to temporal regulation of the progression of viral infection. When expressed alone, outside the context of the virus, E4orf4 induces p53-independent cell-death in transformed cells. Oncogenic transformation of primary cells in tissue culture sensitizes them to cell killing by E4orf4, indicating that E4orf4 research may have implications for cancer therapy. It has also been reported that E4orf4 induces a caspase-independent, non-classical apoptotic pathway, which maintains crosstalk with classical caspase-dependent pathways. Furthermore, several E4orf4 activities in the nucleus and in the cytoplasm and various protein partners contribute to cell killing by this viral protein. In the following chapter I summarize the current knowledge of the unique mode of E4orf4-induced cell death and its underlying mechanisms. Although several explanations for the cancer-specificity of E4orf4-induced toxicity have been proposed, a better grasp of the mechanisms responsible for E4orf4-induced cell death is required to elucidate the differential sensitivity of normal and cancer cells to E4orf4.


Subject(s)
Adenoviridae , Apoptosis/genetics , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Viral Proteins , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
9.
Nucleic Acids Res ; 39(15): 6414-27, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21546548

ABSTRACT

The adenovirus E4 open-reading-frame 4 (E4orf4) protein regulates the progression of viral infection and when expressed individually it induces non-classical apoptosis in transformed cells. Here we show that E4orf4 associates with the ATP-dependent chromatin-remodeling factor ACF that consists of a sucrose non fermenting-2h (SNF2h) ATPase and an Acf1 regulatory subunit. Furthermore, E4orf4 targets protein phosphatase 2A (PP2A) to this complex and to chromatin. Obstruction of SNF2h activity inhibits E4orf4-induced cell death, whereas knockdown of Acf1 results in enhanced E4orf4-induced toxicity in both mammalian and yeast cells, and Acf1 overexpression inhibits E4orf4's ability to downregulate early adenovirus gene expression in the context of viral infection. Knockdown of the Acf1 homolog, WSTF, inhibits E4orf4-induced cell death. Based on these results we suggest that the E4orf4-PP2A complex inhibits ACF and facilitates enhanced chromatin-remodeling activities of other SNF2h-containing complexes, such as WSTF-SNF2h. The resulting switch in chromatin remodeling determines life versus death decisions and contributes to E4orf4 functions during adenovirus infection.


Subject(s)
Adenosine Triphosphatases/metabolism , Apoptosis , Chromosomal Proteins, Non-Histone/metabolism , Protein Phosphatase 2/metabolism , Transcription Factors/metabolism , Viral Proteins/metabolism , Chromatin/metabolism , Chromatin Assembly and Disassembly , Down-Regulation , Fungal Proteins/metabolism , Gene Expression Regulation, Viral , HEK293 Cells , Humans , Transcription Factors/physiology , Viral Proteins/antagonists & inhibitors , Yeasts/metabolism
10.
FEBS Lett ; 594(12): 1891-1917, 2020 06.
Article in English | MEDLINE | ID: mdl-31792953

ABSTRACT

The adenovirus (Ad) early region 4 open reading frame 4 (E4orf4) protein is a small 14-kDa polypeptide endowed with important viral regulatory functions. Although deletion of E4orf4 does not have a major effect on Ad replication due to redundancy among many Ad proteins, E4orf4 provides several functions that improve viral replication. E4orf4 contributes to temporal regulation of virus infection by downregulating early viral gene expression and by altering splicing patterns of Ad mRNAs. It also optimizes the cellular environment for Ad replication by activating the mammalian target of rapamycin pathway to increase viral protein production and by impacting the cell cycle. In addition, E4orf4 participates in the inhibition of the host DNA damage response, promoting the ability of Ad to counteract this antiviral defense mechanism. To fulfill these functions, E4orf4 interacts with numerous cellular proteins, including the major E4orf4 partner, protein phosphatase 2A (PP2A). When expressed alone, outside the context of virus infection, E4orf4 induces an evolutionarily conserved, caspase-independent, cancer-selective cell death with many interesting characteristics. This review critically describes E4orf4's contribution to Ad infection and cancer-cell death.


Subject(s)
Adenoviridae Infections/virology , Neoplasms/pathology , Viral Proteins/chemistry , Viral Proteins/physiology , Adenoviridae Infections/pathology , Alternative Splicing , Animals , Cell Cycle , Cell Death , Cytoplasm/metabolism , DNA Damage , DNA Replication , Gene Expression Regulation, Viral , Host-Pathogen Interactions/physiology , Humans , Neoplasms/virology , Oncolytic Viruses/genetics
11.
Viruses ; 12(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32906746

ABSTRACT

Virus-host cell interactions include several skirmishes between the virus and its host, and the DNA damage response (DDR) network is one of their important battlegrounds. Although some aspects of the DDR are exploited by adenovirus (Ad) to improve virus replication, especially at the early phase of infection, a large body of evidence demonstrates that Ad devotes many of its proteins, including E1B-55K, E4orf3, E4orf4, E4orf6, and core protein VII, and utilizes varied mechanisms to inhibit the DDR. These findings indicate that the DDR would strongly restrict Ad replication if allowed to function efficiently. Various Ad serotypes inactivate DNA damage sensors, including the Mre11-Rad50-Nbs1 (MRN) complex, DNA-dependent protein kinase (DNA-PK), and Poly (ADP-ribose) polymerase 1 (PARP-1). As a result, these viruses inhibit signaling via DDR transducers, such as the ataxia-telangiectasia mutated (ATM) and ATM- and Rad3-related (ATR) kinases, to downstream effectors. The different Ad serotypes utilize both shared and distinct mechanisms to inhibit various branches of the DDR. The aim of this review is to understand the interactions between Ad proteins and the DDR and to appreciate how these interactions contribute to viral replication.


Subject(s)
Adenoviridae Infections/genetics , Adenoviridae Infections/virology , Adenoviridae/physiology , DNA Damage , Adenoviridae/genetics , Adenoviridae Infections/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Humans , Signal Transduction , Virus Replication
12.
J Virol ; 82(19): 9381-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18653458

ABSTRACT

The adenovirus E4 open reading frame 4 (E4orf4) protein is a multifunctional viral regulator that is involved in the temporal regulation of viral gene expression by modulating cellular and viral genes at the transcription and translation levels and by controlling alternative splicing of adenoviral late mRNAs. When expressed individually, E4orf4 induces apoptosis in transformed cells. Using oligonucleotide microarray analysis, validated by quantitative real time PCR, we found that MYC (also known as c-Myc) is downregulated early after the induction of E4orf4 expression. As a result, Myc protein levels are reduced in E4orf4-expressing cells. MYC downregulation is observed both when E4orf4 is expressed individually and within the context of viral infection. E4orf4 reduces MYC transcription but does not affect transcriptional elongation or RNA stability. An interaction with the PP2A-B55 subunit is required for the downregulation of MYC by E4orf4. Since Myc overexpression was previously shown to inhibit adenovirus replication, the downregulation of Myc by E4orf4 would contribute to efficient virus infection.


Subject(s)
Adenoviridae/metabolism , Gene Expression Regulation, Viral , Protein Phosphatase 2/chemistry , Proto-Oncogene Proteins c-myc/metabolism , Viral Proteins/genetics , Viral Proteins/physiology , Adenoviridae/genetics , Cell Cycle , Cell Line , Cell Nucleus/metabolism , Down-Regulation , Exons , Humans , Models, Biological , Mutation , Oligonucleotide Array Sequence Analysis , Open Reading Frames
13.
Cell Death Dis ; 10(6): 455, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186403

ABSTRACT

The adenovirus (Ad) E4orf4 protein contributes to efficient progression of virus infection. When expressed alone E4orf4 induces p53- and caspase-independent cell-death, which is more effective in cancer cells than in normal cells in tissue culture. Cancer selectivity of E4orf4-induced cell-death may result from interference with various regulatory pathways that cancer cells are more dependent on, including DNA damage signaling and proliferation control. E4orf4 signaling is conserved in several organisms, including yeast, Drosophila, and mammalian cells, indicating that E4orf4-induced cell-death can be investigated in these model organisms. The Drosophila genetic model system has contributed significantly to the study of cancer and to identification of novel cancer therapeutics. Here, we used the fly model to investigate the ability of E4orf4 to eliminate cancer tissues in a whole organism with minimal damage to normal tissues. We show that E4orf4 dramatically inhibited tumorigenesis and rescued survival of flies carrying a variety of tumors, including highly aggressive and metastatic tumors in the fly brain and eye discs. Moreover, E4orf4 rescued the morphology of adult eyes containing scrib- cancer clones even when expressed at a much later stage than scrib elimination. The E4orf4 partner protein phosphatase 2A (PP2A) was required for inhibition of tumorigenesis by E4orf4 in the system described here, whereas another E4orf4 partner, Src kinase, provided only minimal contribution to this process. Our results suggest that E4orf4 is an effective anticancer agent and reveal a promising potential for E4orf4-based cancer treatments.


Subject(s)
Drosophila/genetics , Neoplasms, Experimental/therapy , Viral Proteins/metabolism , Animals , Cell Death/genetics , Cell Differentiation/genetics , Disease Models, Animal , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Eye/metabolism , Eye/pathology , Eye Neoplasms/genetics , Eye Neoplasms/metabolism , Eye Neoplasms/mortality , Eye Neoplasms/therapy , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasms, Experimental/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Signal Transduction/genetics , Viral Proteins/genetics , ras Proteins/genetics , ras Proteins/metabolism , ras Proteins/toxicity , src-Family Kinases/genetics , src-Family Kinases/metabolism
14.
Viruses ; 7(5): 2334-57, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25961489

ABSTRACT

During adenovirus (Ad) replication the Ad E4orf4 protein regulates progression from the early to the late phase of infection. However, when E4orf4 is expressed alone outside the context of the virus it induces a non-canonical mode of programmed cell death, which feeds into known cell death pathways such as apoptosis or necrosis, depending on the cell line tested. E4orf4-induced cell death has many interesting and unique features including a higher susceptibility of cancer cells to E4orf4-induced cell killing compared with normal cells, caspase-independence, a high degree of evolutionary conservation of the signaling pathways, a link to perturbations of the cell cycle, and involvement of two distinct cell death programs, in the nucleus and in the cytoplasm. Several E4orf4-interacting proteins including its major partners, protein phosphatase 2A (PP2A) and Src family kinases, contribute to induction of cell death. The various features of E4orf4-induced cell killing as well as studies to decipher the underlying mechanisms are described here. Many explanations for the cancer specificity of E4orf4-induced cell death have been proposed, but a full understanding of the reasons for the different susceptibility of cancer and normal cells to killing by E4orf4 will require a more detailed analysis of the complex E4orf4 signaling network. An improved understanding of the mechanisms involved in this unique mode of programmed cell death may aid in design of novel E4orf4-based cancer therapeutics.


Subject(s)
Adenoviridae/genetics , Apoptosis , Gene Expression , Viral Proteins/genetics , Viral Proteins/metabolism , Cell Line, Tumor , Humans , Necrosis
15.
Front Biosci ; 7: d1369-95, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11991831

ABSTRACT

Adenovirus infection of quiescent cells induces transition from G0 or G1 into the S phase of the cell cycle and allows cellular proliferation. This is beneficial for the virus since cells in S phase provide optimal conditions for viral replication. Adenovirus E1A, E1B and E4 gene products contribute to cell cycle deregulation. E1A proteins inactivate the pRb checkpoint, allowing the E2F transcription factor to activate genes involved in nucleotide metabolism and DNA replication, which are required in S phase. E1A also interacts with transcriptional modulators, including histone acetyltransferases, histone deacetylases, and other chromatin remodeling factors. These interactions affect transcription of several cellular and viral genes, some of which are involved in cell cycle regulation. Cell cycle deregulation by E1A results in stabilization and accumulation of p53. To prevent cell cycle arrest and apoptosis that would be triggered by p53, the adenovirus E1B and E4orf6 gene products employ various mechanisms to inactivate the tumor suppressor. Additional E4 gene products also interact with and modulate cell cycle regulators. Cell cycle checkpoints targeted by adenovirus proteins are often compromised in human tumors as well. Thus, understanding the interactions between adenovirus and the cell cycle has facilitated the generation of adenovirus mutants, which can replicate only in cells with inactivated checkpoints. Such "oncolytic" viruses are being tested for their ability to specifically replicate in and lyse cancer cells.


Subject(s)
Adenoviridae/physiology , Cell Cycle/physiology , Adenoviridae/growth & development , Adenovirus E1A Proteins/physiology , Adenovirus E1B Proteins/physiology , Animals , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Gene Expression Regulation , Humans , Models, Biological , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
16.
Methods Enzymol ; 366: 175-87, 2003.
Article in English | MEDLINE | ID: mdl-14674249

ABSTRACT

The RRS system facilitated the discovery of hitherto unknown interactions with the PP2A-B55 subunit. The advantages of the system lie in its ability to identify interactions that may not be detected by traditional yeast two-hybrid systems. The RRS can thus provide a complementary genetic approach to the identification of protein-protein interactions.


Subject(s)
Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , ras Proteins , Binding Sites , Cell Division , Indicators and Reagents , Phosphoprotein Phosphatases/genetics , Plasmids/genetics , Protein Subunits/chemistry , Protein Subunits/metabolism , Reproducibility of Results , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/chemistry
17.
J Vis Exp ; (68)2012 Oct 21.
Article in English | MEDLINE | ID: mdl-23117279

ABSTRACT

Functional inactivation of gene expression in mammalian cells is crucial for the study of the contribution of a protein of interest to various pathways(1,2). However, conditional knockdown of gene expression is required in cases when constitutive knockdown is not tolerated by cells for a long period of time(3-5). Here we describe a protocol for preparation of cell lines allowing conditional knockdown of subunits of the ACF chromatin remodeling factor. These cell lines facilitate the determination of the contribution of ACF to induction of cell death by the adenovirus E4orf4 protein(6). Sequences encoding short hairpin RNAs for the Acf1 and SNF2h subunits of the ACF chromatin remodeling factor were cloned next to a doxycycline-inducible promoter in a plasmid also containing a gene for the neomycin resistance gene. Neomycin-resistant cell clones were selected in the presence of G418 and isolated. The resulting cell lines were induced by doxycycline treatment, and once Acf1 or SNF2h expression levels were reduced, the cells were transfected with a plasmid encoding E4orf4 or an empty vector. To confirm the specific effect of the shRNA constructs, Acf1 or SNF2h protein levels were restored to WT levels by cotransfection with a plasmid expressing Acf1 or SNF2h which were rendered resistant to the shRNA by introduction of silent mutations. The ability of E4orf4 to induce cell death in the various samples was determined by a DAPI assay, in which the frequency of appearance of nuclei with apoptotic morphologies in the transfected cell population was measured(7-9). The protocol described here can be utilized for determination of the functional contribution of various proteins to induction of cell death by their protein partners in cases when constitutive knockdown may be cell lethal.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Gene Knockdown Techniques/methods , Viral Proteins/genetics , Cell Death/genetics , Cell Line , Chromosomal Proteins, Non-Histone/deficiency , HEK293 Cells , Humans , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transfection/methods
18.
PLoS One ; 5(11): e15539, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-21124936

ABSTRACT

The adenovirus E4 open reading frame 4 (E4orf4) protein contributes to regulation of the progression of virus infection. When expressed individually, E4orf4 was shown to induce non-classical transformed cell-specific apoptosis in mammalian cells. At least some of the mechanisms underlying E4orf4-induced toxicity are conserved from yeast to mammals, including the requirement for an interaction of E4orf4 with protein phosphatase 2A (PP2A). A genetic screen in yeast revealed that the Golgi apyrase Ynd1 associates with E4orf4 and contributes to E4orf4-induced toxicity, independently of Ynd1 apyrase activity. Ynd1 and PP2A were shown to contribute additively to E4orf4-induced toxicity in yeast, and to interact genetically and physically. A mammalian orthologue of Ynd1 was shown to bind E4orf4 in mammalian cells, confirming the evolutionary conservation of this interaction. Here, we use mutation analysis to identify the cytosolic tail of Ynd1 as the protein domain required for mediation of the E4orf4 toxic signal and for the interaction with E4orf4. We also show that E4orf4 associates with cellular membranes in yeast and is localized at their cytoplasmic face. However, E4orf4 is membrane-associated even in the absence of Ynd1, suggesting that additional membrane proteins may mediate E4orf4 localization. Based on our results and on a previous report describing a collection of Ynd1 protein partners, we propose that the Ynd1 cytoplasmic tail acts as a scaffold, interacting with a multi-protein complex, whose targeting by E4orf4 leads to cell death.


Subject(s)
Apyrase/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Viral Proteins/metabolism , Adenovirus E4 Proteins/genetics , Adenovirus E4 Proteins/metabolism , Apyrase/genetics , Binding Sites/genetics , Golgi Apparatus/metabolism , Immunoblotting , Immunoprecipitation , Mutagenesis , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Viral Proteins/genetics
20.
J Biol Chem ; 280(50): 41270-7, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16227198

ABSTRACT

Adenovirus E4orf4 (early region 4 open reading frame 4) protein induces protein phosphatase 2A-dependent non-classical apoptosis in mammalian cells and irreversible growth arrest in Saccharomyces cerevisiae. Oncogenic transformation sensitizes cells to E4orf4-induced cell death. To uncover additional components of the E4orf4 network required for induction of its unique mode of apoptosis, we used yeast genetics to select gene deletions conferring resistance to E4orf4. Deletion of YND1, encoding a yeast Golgi apyrase, conferred partial resistance to E4orf4. However, Ynd1p apyrase activity was not required for E4orf4-induced toxicity. Ynd1p and Cdc55p, the yeast protein phosphatase 2A-B subunit, contributed additively to E4orf4-induced toxicity. Furthermore, concomitant overexpression of one and deletion of the other was detrimental to yeast growth, demonstrating a functional interaction between the two proteins. YND1 and CDC55 also interacted genetically with CDC20 and CDH1/HCT1, encoding activating subunits of the anaphase-promoting complex/cyclosome. In addition to their functional interaction, Ynd1p and Cdc55p interacted physically, and this interaction was disrupted by E4orf4, which remained associated with both proteins. The results suggested that Ynd1p and Cdc55p share a common downstream target whose balanced modulation by the two E4orf4 partners is crucial to viability. Disruption of this balance by E4orf4 may lead to cell death. NTPDase-4/Lalp70/UDPase, the closest mammalian homologue of Ynd1p, associated with E4orf4 in mammalian cells, suggesting that the results in yeast are relevant to the mammalian system.


Subject(s)
Apyrase/metabolism , Cell Cycle Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Adenovirus E4 Proteins , Anaphase , Apoptosis , Cdc20 Proteins , Cell Death , Cell Line , Cell Transformation, Neoplastic , DNA Transposable Elements , Gene Deletion , Gene Expression Regulation, Fungal , Genetic Vectors , Genotype , Golgi Apparatus/metabolism , Humans , Image Processing, Computer-Assisted , Immunoblotting , Immunoprecipitation , Mutation , Open Reading Frames , Plasmids/metabolism , Protein Binding , Protein Phosphatase 2 , Saccharomyces cerevisiae/metabolism , Signal Transduction , Spindle Apparatus
SELECTION OF CITATIONS
SEARCH DETAIL