Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Biomed Pharmacother ; 168: 115635, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37816303

ABSTRACT

Primary and acquired therapy resistance is a major problem in patients with BRAF-mutant melanomas being treated with BRAF and MEK inhibitors (BRAFI, MEKi). Therefore, development of alternative therapy regimes is still required. In this regard, new drug combinations targeting different pathways to induce apoptosis could offer promising alternative approaches. Here, we investigated the combination of proteasome and Kv1.3 potassium channel inhibition on chemo-resistant, BRAF inhibitor-resistant as well as sensitive human melanoma cells. Our experiments demonstrated that all analyzed melanoma cell lines were sensitive to proteasome inhibitor treatment at concentrations that are not toxic to primary human fibroblasts. To further reduce proteasome inhibitor-associated side effects, and to foster apoptosis, potassium channels, which are other targets to induce pro-apoptotic effects in cancer cells, were blocked. In support, combined exposure of melanoma cells to proteasome and Kv1.3 channel inhibitor resulted in synergistic effects and significantly reduced cell viability. On the molecular level, enhanced apoptosis correlated with an increase of intracellular Kv1.3 channels and pro-apoptotic proteins such as Noxa and Bak and a reduction of anti-apoptotic proteins. Thus, use of combined therapeutic strategies triggering different apoptotic pathways may efficiently prevent the outgrowth of drug-resistant and -sensitive BRAF-mutant melanoma cells. In addition, this could be the basis for an alternative approach to treat other tumors expressing mutated BRAF such as non-small-cell lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Melanoma , Humans , Proteasome Endopeptidase Complex/metabolism , Kv1.3 Potassium Channel/genetics , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm , Cell Line, Tumor , Lung Neoplasms/drug therapy , Melanoma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Apoptosis Regulatory Proteins/metabolism , Mutation
2.
PLoS Pathog ; 18(1): e1010193, 2022 01.
Article in English | MEDLINE | ID: mdl-34982803

ABSTRACT

The chimpanzee cytomegalovirus (CCMV) is the closest relative of human CMV (HCMV). Because of the high conservation between these two species and the ability of human cells to fully support CCMV replication, CCMV holds great potential as a model system for HCMV. To make the CCMV genome available for precise and rapid gene manipulation techniques, we captured the genomic DNA of CCMV strain Heberling as a bacterial artificial chromosome (BAC). Selected BAC clones were reconstituted to infectious viruses, growing to similar high titers as parental CCMV. DNA sequencing confirmed the integrity of our clones and led to the identification of two polymorphic loci and a deletion-prone region within the CCMV genome. To re-evaluate the CCMV coding potential, we analyzed the viral transcriptome and proteome and identified several novel ORFs, splice variants, and regulatory RNAs. We further characterized the dynamics of CCMV gene expression and found that viral proteins cluster into five distinct temporal classes. In addition, our datasets revealed that the host response to CCMV infection and the de-regulation of cellular pathways are in line with known hallmarks of HCMV infection. In a first functional experiment, we investigated a proposed frameshift mutation in UL128 that was suspected to restrict CCMV's cell tropism. In fact, repair of this frameshift re-established productive CCMV infection in endothelial and epithelial cells, expanding the options of CCMV as an infection model. Thus, BAC-cloned CCMV can serve as a powerful tool for systematic approaches in comparative functional genomics, exploiting the close phylogenetic relationship between CCMV and HCMV.


Subject(s)
Cytomegalovirus/genetics , Pan troglodytes/virology , Animals , Cytomegalovirus Infections/virology , Disease Models, Animal , Genome, Viral , Humans
3.
Nat Commun ; 12(1): 3582, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34117224

ABSTRACT

In mouse development, long-term silencing by CpG island DNA methylation is specifically targeted to germline genes; however, the molecular mechanisms of this specificity remain unclear. Here, we demonstrate that the transcription factor E2F6, a member of the polycomb repressive complex 1.6 (PRC1.6), is critical to target and initiate epigenetic silencing at germline genes in early embryogenesis. Genome-wide, E2F6 binds preferentially to CpG islands in embryonic cells. E2F6 cooperates with MGA to silence a subgroup of germline genes in mouse embryonic stem cells and in embryos, a function that critically depends on the E2F6 marked box domain. Inactivation of E2f6 leads to a failure to deposit CpG island DNA methylation at these genes during implantation. Furthermore, E2F6 is required to initiate epigenetic silencing in early embryonic cells but becomes dispensable for the maintenance in differentiated cells. Our findings elucidate the mechanisms of epigenetic targeting of germline genes and provide a paradigm for how transient repression signals by DNA-binding factors in early embryonic cells are translated into long-term epigenetic silencing during mouse development.


Subject(s)
CpG Islands/genetics , E2F6 Transcription Factor/genetics , E2F6 Transcription Factor/metabolism , Embryonic Development/genetics , Epigenesis, Genetic , Germ Cells/metabolism , Animals , Binding Sites , CRISPR-Cas Systems , Cell Differentiation , DNA Methylation , Gene Silencing , Mice , Mice, Knockout , Mouse Embryonic Stem Cells , Polycomb Repressive Complex 1/metabolism , RNA, Small Interfering
4.
Nat Commun ; 11(1): 4845, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32973148

ABSTRACT

Herpesviruses encode conserved protein kinases (CHPKs) to stimulate phosphorylation-sensitive processes during infection. How CHPKs bind to cellular factors and how this impacts their regulatory functions is poorly understood. Here, we use quantitative proteomics to determine cellular interaction partners of human herpesvirus (HHV) CHPKs. We find that CHPKs can target key regulators of transcription and replication. The interaction with Cyclin A and associated factors is identified as a signature of ß-herpesvirus kinases. Cyclin A is recruited via RXL motifs that overlap with nuclear localization signals (NLS) in the non-catalytic N termini. This architecture is conserved in HHV6, HHV7 and rodent cytomegaloviruses. Cyclin A binding competes with NLS function, enabling dynamic changes in CHPK localization and substrate phosphorylation. The cytomegalovirus kinase M97 sequesters Cyclin A in the cytosol, which is essential for viral inhibition of cellular replication. Our data highlight a fine-tuned and physiologically important interplay between a cellular cyclin and viral kinases.


Subject(s)
DNA Replication/physiology , Herpesviridae Infections/metabolism , Herpesviridae/metabolism , Protein Kinases/metabolism , Animals , Cyclin A/genetics , Cyclin A/metabolism , Cytomegalovirus/genetics , DNA/metabolism , HEK293 Cells , Herpesviridae/enzymology , Herpesviridae/genetics , Herpesviridae Infections/virology , Humans , Mice , NIH 3T3 Cells , Nuclear Localization Signals/metabolism , Phosphorylation , Protein Interaction Maps , Viral Proteins/genetics , Viral Proteins/metabolism
5.
PLoS Pathog ; 13(1): e1006193, 2017 01.
Article in English | MEDLINE | ID: mdl-28129404

ABSTRACT

Generally, the antagonism between host restriction factors and viral countermeasures decides on cellular permissiveness or resistance to virus infection. Human cytomegalovirus (HCMV) has evolved an additional level of self-imposed restriction by the viral tegument protein pp150. Depending on a cyclin A-binding motif, pp150 prevents the onset of viral gene expression in the S/G2 cell cycle phase of otherwise fully permissive cells. Here we address the physiological relevance of this restriction during productive HCMV infection by employing a cyclin A-binding deficient pp150 mutant virus. One consequence of unrestricted viral gene expression in S/G2 was the induction of a G2/M arrest. G2-arrested but not mitotic cells supported viral replication. Cyclin A destabilization by the viral gene product pUL21a was required to maintain the virus-permissive G2-arrest. An HCMV double-point mutant where both pp150 and pUL21a are disabled in cyclin A interaction forced mitotic entry of the majority of infected cells, with a severe negative impact on cell viability and virus growth. Thus, pp150 and pUL21a functionally cooperate, together building a cell cycle synchronization strategy of cyclin A targeting and avoidance that is essential for productive HCMV infection.


Subject(s)
Cyclin A/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus/pathogenicity , Phosphoproteins/metabolism , Synthetic Lethal Mutations/genetics , Viral Matrix Proteins/metabolism , Virus Replication/physiology , Cells, Cultured , Cytomegalovirus/metabolism , Cytomegalovirus Infections/metabolism , Flow Cytometry , Host-Pathogen Interactions/genetics , Humans , Immunoblotting
6.
Target Oncol ; 10(4): 523-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25559288

ABSTRACT

After extensive research on radiochemotherapy, 5-year survival rates of children with high risk neuroblastoma still do not exceed 50%, owing to adverse side-effects exemplified by doxorubicin-induced cardiomyopathy. A promising new approach is the combination of conventional therapies with specific modulation of cell signaling pathways promoting therapeutic resistance, such as inhibition of aberrant kinase activity or re-expression of silenced tumor suppressor genes by means of chromatin remodeling. In this regard, we established a system that allows to identify potential drug targets as well as to validate respective candidate inhibitors in high-risk neuroblastoma model cell lines. Cell culture, drug exposure, shRNA-mediated knockdown and phenotype analysis are integrated into an efficient and versatile single well-based protocol. By utilizing this system, we assessed RG108, SGI-1027 and nanaomycin A, three novel DNA methyltransferase inhibitors that have not been tested in neuroblastoma cell lines so far, for their potential of synergistic anti-tumor activity in combination with doxorubicin. We found that, similarly to azacytidine, SGI-1027 and nanaomycin A mediate synergistic growth inhibition with doxorubicin independently of N-Myc status. However, they display high cytotoxicity but lack global DNA demethylation activity. Secondly, we conducted a lentiviral shRNA screen of F-box proteins, key regulators of protein stability, and identified Fbxw11/ß-TrCP2 as well as Fbxo5/Emi1 as potential therapeutic targets in neuroblastoma. These results complement existing studies and underline the reliability and versatility of our single well-based protocol.


Subject(s)
DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , F-Box Proteins/genetics , Neuroblastoma/therapy , Aminoquinolines/administration & dosage , Aminoquinolines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Synergism , Enzyme Inhibitors/administration & dosage , HEK293 Cells , Humans , Molecular Targeted Therapy , Naphthoquinones/administration & dosage , Naphthoquinones/pharmacology , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/pathology , Phthalimides/administration & dosage , Phthalimides/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Tryptophan/administration & dosage , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
7.
PLoS Pathog ; 10(10): e1004514, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25393019

ABSTRACT

Entry into mitosis is accompanied by dramatic changes in cellular architecture, metabolism and gene expression. Many viruses have evolved cell cycle arrest strategies to prevent mitotic entry, presumably to ensure sustained, uninterrupted viral replication. Here we show for human cytomegalovirus (HCMV) what happens if the viral cell cycle arrest mechanism is disabled and cells engaged in viral replication enter into unscheduled mitosis. We made use of an HCMV mutant that, due to a defective Cyclin A2 binding motif in its UL21a gene product (pUL21a), has lost its ability to down-regulate Cyclin A2 and, therefore, to arrest cells at the G1/S transition. Cyclin A2 up-regulation in infected cells not only triggered the onset of cellular DNA synthesis, but also promoted the accumulation and nuclear translocation of Cyclin B1-CDK1, premature chromatin condensation and mitotic entry. The infected cells were able to enter metaphase as shown by nuclear lamina disassembly and, often irregular, metaphase spindle formation. However, anaphase onset was blocked by the still intact anaphase promoting complex/cyclosome (APC/C) inhibitory function of pUL21a. Remarkably, the essential viral IE2, but not the related chromosome-associated IE1 protein, disappeared upon mitotic entry, suggesting an inherent instability of IE2 under mitotic conditions. Viral DNA synthesis was impaired in mitosis, as demonstrated by the abnormal morphology and strongly reduced BrdU incorporation rates of viral replication compartments. The prolonged metaphase arrest in infected cells coincided with precocious sister chromatid separation and progressive fragmentation of the chromosomal material. We conclude that the Cyclin A2-binding function of pUL21a contributes to the maintenance of a cell cycle state conducive for the completion of the HCMV replication cycle. Unscheduled mitotic entry during the course of the HCMV replication has fatal consequences, leading to abortive infection and cell death.


Subject(s)
Cyclin A2/metabolism , Cytomegalovirus/physiology , DNA Replication , Viral Proteins/metabolism , Virus Replication , Cell Cycle , Cell Cycle Checkpoints , Cell Line , Cyclin A2/genetics , Cytomegalovirus/genetics , Gene Expression Regulation , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mitosis , Proteasome Endopeptidase Complex , Protein Interaction Mapping , Trans-Activators/genetics , Trans-Activators/metabolism , Up-Regulation , Viral Proteins/genetics
8.
Methods Mol Biol ; 1119: 123-32, 2014.
Article in English | MEDLINE | ID: mdl-24639222

ABSTRACT

The cell cycle position at the time of infection has a profound influence on human cytomegalovirus (HCMV) gene expression and therefore needs consideration in the design and control of HCMV experiments. While G0/G1 cells support the immediate onset of viral transcription, cells progressing through the S and G2 cell cycle phases prevent HCMV from entering the lytic replication cycle. Here, we provide two fast and reliable protocols that allow one to determine the cell cycle distribution of the designated host cells and monitor viral protein expression as a function of the cell cycle state. Both protocols make use of the thymidine analogue 5-ethynyl-2'-deoxyuridine and "click" chemistry to label HCMV-non-permissive S phase cells in a gentle and sensitive way.


Subject(s)
Cytomegalovirus/genetics , Molecular Biology/methods , Viral Proteins/biosynthesis , Virus Replication/genetics , Click Chemistry , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Deoxyuridine/analogs & derivatives , Deoxyuridine/chemistry , Flow Cytometry , Gene Expression Regulation, Viral , Humans
9.
Proc Natl Acad Sci U S A ; 110(43): 17510-5, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24101496

ABSTRACT

Upon cell entry, herpesviruses deliver a multitude of premade virion proteins to their hosts. The interplay between these incoming proteins and cell-specific regulatory factors dictates the outcome of infections at the cellular level. Here, we report a unique type of virion-host cell interaction that is essential for the cell cycle and differentiation state-dependent onset of human cytomegalovirus (HCMV) lytic gene expression. The major tegument 150-kDa phosphoprotein (pp150) of HCMV binds to cyclin A2 via a functional RXL/Cy motif resulting in its cyclin A2-dependent phosphorylation. Alanine substitution of the RXL/Cy motif prevents this interaction and allows the virus to fully escape the cyclin-dependent kinase (CDK)-mediated block of immediate early (IE) gene expression in S/G2 phase that normally restricts the onset of the HCMV replication cycle to G0/G1. Furthermore, the cyclin A2-CDK-pp150 axis is also involved in the establishment of HCMV quiescence in NTera2 cells, showing the importance of this molecular switch for differentiation state-dependent regulation of IE gene expression. Consistent with the known nucleocapsid-binding function of pp150, its RXL/Cy-dependent phosphorylation affects gene expression of the parental virion only, suggesting a cis-acting, virus particle-associated mechanism of control. The pp150 homologs of other primate and mammalian CMVs lack an RXL/Cy motif and accordingly even the nearest relative of HCMV, chimpanzee CMV, starts its lytic cycle in a cell cycle-independent manner. Thus, HCMV has evolved a molecular sensor for cyclin A2-CDK activity to restrict its IE gene expression program as a unique level of self-limitation and adaptation to its human host.


Subject(s)
Cell Cycle , Cell Differentiation , Cyclin A2/metabolism , Cyclin-Dependent Kinases/metabolism , Cytomegalovirus/metabolism , Phosphoproteins/metabolism , Viral Matrix Proteins/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Cyclin A2/genetics , Cyclin-Dependent Kinases/genetics , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Flow Cytometry , Gene Expression Regulation, Viral , Genes, Immediate-Early/genetics , HEK293 Cells , Host-Pathogen Interactions , Humans , Immunoblotting , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Mutation , Phosphoproteins/genetics , Phosphorylation , Protein Binding , Viral Matrix Proteins/genetics
10.
J Virol ; 86(17): 9369-83, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22718829

ABSTRACT

Human cytomegalovirus (HCMV) starts its lytic replication cycle only in the G(0)/G(1) phase of the cell division cycle. S/G(2) cells can be infected but block the onset of immediate-early (IE) gene expression. This block can be overcome by inhibition of cyclin-dependent kinases (CDKs), suggesting that cyclin A2, the only cyclin with an S/G(2)-specific activity profile, may act as a negative regulator of viral gene expression. To directly test this hypothesis, we generated derivatives of an HCMV-permissive glioblastoma cell line that express cyclin A2 in a constitutive, cell cycle-independent manner. We demonstrate that even moderate cyclin A2 overexpression in G(1) was sufficient to severely compromise the HCMV replicative cycle after high-multiplicity infection. This negative effect was composed of a strong but transient inhibition of IE gene transcription and a more sustained alteration of IE mRNA processing, resulting in reduced levels of UL37 and IE2, an essential transactivator of viral early gene expression. Consistently, cyclin A2-overexpressing cells showed a strong delay of viral early and late gene expression, as well as virus reproduction. All effects were dependent on CDK activity, as a cyclin A2 mutant deficient in CDK binding was unable to interfere with the HCMV infectious cycle. Interestingly, murine CMV, whose IE gene expression is known to be cell cycle independent, is not affected by cyclin A2. Instead, it upregulates cyclin A2-associated kinase activity upon infection. Understanding the mechanisms behind the HCMV-specific action of cyclin A2-CDK might reveal new targets for antiviral strategies.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin A2/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cytomegalovirus Infections/enzymology , Cytomegalovirus/genetics , Down-Regulation , Immediate-Early Proteins/metabolism , Trans-Activators/metabolism , Animals , CDC2 Protein Kinase/genetics , Cell Cycle , Cell Line, Tumor , Cyclin A2/genetics , Cyclin-Dependent Kinase 2/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Gene Expression Regulation, Viral , Humans , Immediate-Early Proteins/genetics , Mice , Trans-Activators/genetics
11.
Angew Chem Int Ed Engl ; 51(26): 6516-20, 2012 Jun 25.
Article in English | MEDLINE | ID: mdl-22644704

ABSTRACT

Eraserhead: Stem cells seem to erase epigenetic information by decarboxylation of the newly discovered epigenetic base 5-carboxycytosine (caC; see picture). This reaction is likely to involve a nucleophilic attack of the C5-C6 double bond.


Subject(s)
Cytosine/analogs & derivatives , Embryonic Stem Cells/chemistry , Animals , Base Sequence , Cytosine/chemistry , Cytosine/metabolism , DNA/chemistry , Decarboxylation , Embryonic Stem Cells/metabolism , Mice , Nitrogen Isotopes/chemistry , Oxidation-Reduction
12.
J Virol ; 85(21): 11409-21, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21880774

ABSTRACT

Many viruses antagonize tumor necrosis factor alpha (TNF-α) signaling in order to counteract its antiviral properties. One way viruses achieve this goal is to reduce TNF-α receptor 1 (TNFR1) on the surface of infected cells. Such a mechanism is also employed by human cytomegalovirus (HCMV), as recently reported by others and us. On the other hand, TNF-α has also been shown to foster reactivation of HCMV from latency. By characterizing a new variant of HCMV AD169, we show here that TNFR1 downregulation by HCMV only becomes apparent upon infection of cells with HCMV strains lacking the so-called ULb' region. This region contains genes involved in regulating viral immune escape, cell tropism, or latency and is typically lost from laboratory strains but present in low-passage strains and clinical isolates. We further show that although ULb'-positive viruses also contain the TNFR1-antagonizing function, this activity is masked by a dominant TNFR1 upregulation mediated by the ULb' gene product UL138. Isolated expression of UL138 in the absence of viral infection upregulates TNFR1 surface expression and can rescue both TNFR1 reexpression and TNF-α responsiveness of cells infected with an HCMV mutant lacking the UL138-containing transcription unit. Given that the UL138 gene product is one of the few genes recognized to be expressed during HCMV latency and the known positive effects of TNF-α on viral reactivation, we suggest that via upregulating TNFR1 surface expression UL138 may sensitize latently infected cells to TNF-α-mediated reactivation of HCMV.


Subject(s)
Cytomegalovirus/immunology , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Tumor Necrosis Factor-alpha/immunology , Viral Proteins/metabolism , Cells, Cultured , Gene Expression Profiling , Humans , Microarray Analysis
13.
J Gen Virol ; 92(Pt 12): 2757-2769, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21832009

ABSTRACT

The onset of human cytomegalovirus (HCMV) lytic replication is strictly controlled by the host cell division cycle. Although viral entry of S/G2-phase cells is unperturbed expression of major immediate-early (MIE) genes IE1 and IE2 is tightly blocked in these cells. Besides the finding that cyclin-dependent kinase (CDK) activity is required for IE1/IE2 repression little is known about the nature of this cell cycle-dependent block. Here, we show that the block occurs after nuclear entry of viral DNA and prevents the accumulation of IE1/IE2 mRNAs, suggesting an inhibition of transcription. Remarkably, the presence of cis-regulatory regions of the MIE locus is neither sufficient nor necessary for IE1/IE2 repression in the S/G2 phase. Furthermore, the block of viral mRNA expression also affects other immediate-early transcribed regions, i.e. the US3 and UL36-38 gene loci. This suggests a mechanism of repression that acts in a general and not a gene-specific fashion. Such a nuclear, genome-wide repression of HCMV is typically mediated by the intrinsic immune defence at nuclear domain 10 (ND10) structures. However, we found that neither Daxx nor PML, the main players of ND10-based immunity, are required for the block to viral gene expression in the S/G2 phase. In addition, the viral tegument protein pp71 (pUL82), a major antagonist of the intrinsic immunity at pre-immediate-early times of infection, proved to be functional in S-phase cells. This suggests the existence of a yet undiscovered, CDK-dependent mechanism exerting higher-level control over immediate-early mRNA expression in HCMV-infected cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cytomegalovirus/genetics , Gene Expression Regulation, Viral , Genes, Immediate-Early , Nuclear Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , 3T3 Cells , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Cloning, Molecular , Co-Repressor Proteins , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cytomegalovirus/metabolism , Cytomegalovirus/physiology , DNA, Viral/genetics , DNA, Viral/metabolism , G2 Phase , Genetic Loci , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mice , Molecular Chaperones , Mutagenesis , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , RNA, Messenger/genetics , RNA, Messenger/metabolism , S Phase , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Viral Proteins/metabolism , Virus Replication
15.
J Clin Oncol ; 29(23): 3185-93, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21747090

ABSTRACT

PURPOSE: In the clinical management of children with relapsed acute lymphoblastic leukemia (ALL), treatment resistance remains a major challenge. Alterations of the TP53 gene are frequently associated with resistance to chemotherapy, but their significance in relapsed childhood ALL has remained controversial because of small studies. PATIENTS AND METHODS: Therefore, we systematically studied 265 first-relapse patients enrolled in the German Acute Lymphoblastic Leukemia Relapse Berlin-Frankfurt-Mü nster 2002 (ALL-REZ BFM 2002) trial for sequence and copy number alterations of the TP53 gene by using direct sequencing and multiplex ligation-dependent probe amplification. RESULTS: We observed copy number and sequence alterations of TP53 in 12.4% (27 of 218) of patients with B-cell precursor ALL and 6.4% (three of 47) of patients with T-cell ALL relapse. Backtracking to initial ALL in 23 matched samples revealed that 54% of all TP53 alterations were gained at relapse. Within B-cell precursor ALL, TP53 alterations were consistently associated with nonresponse to chemotherapy (P < .001) and poor event-free survival (P < .001) and overall survival rates (P = .002). TP53 alterations also had a significant impact on survival within intermediate-risk (S2) and high-risk (S3/S4) relapse patients (P = .007 and P = .019, respectively). This prognostic significance of TP53 alterations was confirmed in multivariate analysis. Besides their clinical impact, TP53 alterations were associated with a higher fraction of leukemic cells in S/G(2)-M phase of the cell cycle at relapse diagnosis. CONCLUSION: Alterations of the TP53 gene are of particular importance in the relapse stage of childhood ALL, in which they independently predict high risk of treatment failure in a significant number of patients. Therefore, they will aid in future risk assessment of children with ALL relapse.


Subject(s)
Drug Resistance, Neoplasm/genetics , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Tumor Suppressor Protein p53/genetics , Child , Child, Preschool , Clinical Trials as Topic , Disease-Free Survival , Female , Flow Cytometry , Gene Deletion , Humans , Kaplan-Meier Estimate , Male , Multicenter Studies as Topic , Polymerase Chain Reaction , Proportional Hazards Models , Risk Factors , Treatment Outcome
16.
PLoS Pathog ; 6(9): e1001096, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20844576

ABSTRACT

The onset of human cytomegalovirus (HCMV) lytic infection is strictly synchronized with the host cell cycle. Infected G0/G1 cells support viral immediate early (IE) gene expression and proceed to the G1/S boundary where they finally arrest. In contrast, S/G2 cells can be infected but effectively block IE gene expression and this inhibition is not relieved until host cells have divided and reentered G1. During latent infection IE gene expression is also inhibited, and for reactivation to occur this block to IE gene expression must be overcome. It is only poorly understood which viral and/or cellular activities maintain the block to cell cycle or latency-associated viral IE gene repression and whether the two mechanisms may be linked. Here, we show that the block to IE gene expression during S and G2 phase can be overcome by both genotoxic stress and chemical inhibitors of cellular DNA replication, pointing to the involvement of checkpoint-dependent signaling pathways in controlling IE gene repression. Checkpoint-dependent rescue of IE expression strictly requires p53 and in the absence of checkpoint activation is mimicked by proteasomal inhibition in a p53 dependent manner. Requirement for the cyclin dependent kinase (CDK) inhibitor p21 downstream of p53 suggests a pivotal role for CDKs in controlling IE gene repression in S/G2 and treatment of S/G2 cells with the CDK inhibitor roscovitine alleviates IE repression independently of p53. Importantly, CDK inhibiton also overcomes the block to IE expression during quiescent infection of NTera2 (NT2) cells. Thus, a timely block to CDK activity not only secures phase specificity of the cell cycle dependent HCMV IE gene expression program, but in addition plays a hitherto unrecognized role in preventing the establishment of a latent-like state.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Cytomegalovirus/physiology , Immediate-Early Proteins/metabolism , Virus Activation , Virus Latency , Virus Replication , Antibiotics, Antineoplastic/pharmacology , Blotting, Western , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , DNA Replication/drug effects , DNA Replication/radiation effects , Doxorubicin/pharmacology , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immediate-Early Proteins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays
17.
Blood ; 115(18): 3763-71, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20228269

ABSTRACT

A consistently increased mRNA expression of the adhesion receptor CD11b is a hallmark of the reported genomewide gene expression changes in precursor B-cell acute lymphoblastic leukemia (PBC-ALL) after 1 week of induction therapy. To investigate its clinical relevance, CD11b protein expression in leukemic blasts has been prospectively measured at diagnosis (159 patients) and during therapy (53 patients). The initially heterogeneous expression of CD11b inversely correlated with cytoreduction rates measured at clinically significant time points of induction therapy in the ALL-Berlin-Frankfurt-Münster 2000 protocol. CD11b positivity conferred a 5-fold increased risk of minimal residual disease (MRD) after induction therapy (day 33) and of high-risk group assignment after consolidation therapy (day 78). In the multivariate analysis CD11b expression was an independent prognostic factor compared with other clinically relevant parameters at diagnosis. During therapy, CD11b expression increased early in most ALL cases and remained consistently increased during induction/consolidation therapy. In more than 30% of MRD-positive cases, the CD11b expression on blast cells exceeded that of mature memory B cells and improved the discrimination of residual leukemic cells from regenerating bone marrow. Taken together, CD11b expression has considerable implications for prognosis, treatment response monitoring, and MRD detection in childhood PBC-ALL.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , CD11b Antigen/metabolism , Drug Resistance, Neoplasm , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Adolescent , B-Lymphocytes/metabolism , Bone Marrow/metabolism , Child , Child, Preschool , Female , Gene Expression Profiling , Humans , Infant , Male , Neoplasm, Residual , Oligonucleotide Array Sequence Analysis , Prospective Studies , RNA, Messenger/genetics , RNA, Messenger/metabolism , Remission Induction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Treatment Outcome
18.
J Virol ; 82(20): 10188-98, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18667506

ABSTRACT

The infectious cycle of human cytomegalovirus (HCMV) is intricately linked to the host's cell cycle. Viral gene expression can be initiated only in G(0)/G(1) phase. Once expressed, the immediate-early gene product IE2 prevents cellular DNA synthesis, arresting infected cells with a G(1) DNA content. This function is required for efficient viral replication in vitro. A prerequisite for addressing its in vivo relevance is the characterization of cell cycle-regulatory activities of CMV species for which animal models have been established. Here, we show that murine CMV (MCMV), like HCMV, has a strong antiproliferative capacity and arrests cells in G(1). Unexpectedly, and in contrast to HCMV, MCMV can also block cells that have passed through S phase by arresting them in G(2). Moreover, MCMV can also replicate in G(2) cells. This is made possible by the cell cycle-independent expression of MCMV immediate-early genes. Transfection experiments show that of several MCMV candidate genes, only immediate-early gene 3 (ie3), the homologue of HCMV IE2, exhibits cell cycle arrest activity. Accordingly, an MCMV ie3 deletion mutant has lost the ability to arrest cells in either G(1) or G(2). Thus, despite interspecies variations in the cell cycle dependence of viral gene expression, the central theme of HCMV IE2-induced cell cycle arrest is conserved in the murine counterpart, raising the possibility of studying its physiological relevance at the level of the whole organism.


Subject(s)
G1 Phase/physiology , G2 Phase/physiology , Gene Expression Regulation, Viral , Genes, Immediate-Early/genetics , Muromegalovirus/genetics , Muromegalovirus/physiology , Animals , Cell Proliferation , DNA Replication , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mice , Muromegalovirus/pathogenicity , NIH 3T3 Cells , Trans-Activators/genetics , Trans-Activators/metabolism
19.
J Neurooncol ; 86(1): 23-30, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17763999

ABSTRACT

Impaired tumor suppressor functions, such as deficient p53, are characteristic for glioblastoma multiforme (GBM) and can cause resistance to DNA-damaging agents like cisplatin. We have recently shown that the INhibitor of Growth 1 (ING1) tumor suppressor is down-regulated in malignant gliomas and that the decrease of ING1 expression correlates with histological grade of malignancy, suggesting a role for ING1 in the pathogenesis and progression of malignant gliomas. Based on this background, the purpose of our current study was to examine the potential impact of ING1 protein levels on DNA-damage response in GBM. Using LN229 GBM cells, which express ING1 proteins and harbor mutant TP53, we are the first to show that DNA damage by cisplatin or ionizing radiation differentially induced the two major ING1 splicing isoforms. The p47 ING1a isoform, that promotes deacetylation of histones, thus formation of heterochromatic regions of DNA, which are less susceptible to DNA damage, was preferentially induced by >50-fold. This might represent a response to protect DNA from damage. Also, ING1 knockdown by siRNA accelerated transit of cells through G1 phase, consistent with ING1 serving a tumor suppressor function, and caused cells to enter apoptosis more rapidly in response to cisplatin. Our results indicate that malignant gliomas may down-regulate ING1 to allow more efficient tumor growth and progression. Also, ING1 down-regulation may sensitize GBM cells with deficient p53 to treatment with cisplatin.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Down-Regulation/genetics , Glioblastoma , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Proteins/metabolism , Bromodeoxyuridine/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/radiation effects , Down-Regulation/drug effects , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/physiopathology , Humans , Inhibitor of Growth Protein 1 , RNA, Small Interfering/pharmacology , Radiation , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics
20.
J Virol ; 80(23): 11686-98, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17005669

ABSTRACT

NF-kappaB plays an important role in the early cellular response to pathogens by activating genes involved in inflammation, immune response, and cell proliferation and survival. NF-kappaB is also utilized by many viral pathogens, like human cytomegalovirus (HCMV), to activate their own gene expression programs, reflecting intricate roles for NF-kappaB in both antiviral defense mechanisms and viral physiology. Here we show that the NF-kappaB signaling pathway stimulated by proinflammatory cytokines tumor necrosis factor alpha (TNF-alpha) and interleukin-1beta (IL-1beta) becomes inhibited in HCMV-infected cells. The block to NF-kappaB signaling is first noticeable during the early phase of infection but is fully established only at later times. Biochemical and genetic evidence demonstrates that the viral inhibition of proinflammatory signaling by distinct cytokines occurs upstream of the convergence point of NF-kappaB-activating pathways, i.e., the IkappaB kinase complex, and that it is mediated via different mechanisms. Consistent with this, we further show that an HCMV variant that has lost the ability to downregulate TNF-alpha-induced NF-kappaB signaling also fails to downregulate surface expression of TNF receptor 1, thereby mechanistically linking the inhibition of TNF-alpha-induced NF-kappaB signaling by HCMV to TNF receptor targeting. Our data support a model whereby HCMV inhibits cytokine-induced NF-kappaB signaling at later times during infection, and we suggest that this contributes to the inhibition of the cell's antiviral defense program.


Subject(s)
Cytomegalovirus/chemistry , Cytomegalovirus/physiology , Interleukin-1beta/antagonists & inhibitors , NF-kappa B/biosynthesis , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Humans , Interleukin-1beta/metabolism , Protein Serine-Threonine Kinases , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...