19 results on '"Øster, B"'
Search Results
2. Human Herpesvirus 6B Induces Phenotypic Maturation Without IL-10 and IL-12p70 Production in Dendritic Cells
- Author
-
Bertelsen, L. B., Petersen, C. C., Kofod-Olsen, E., Øster, B., Höllsberg, P., Agger, R., and Hokland, M.
- Published
- 2010
- Full Text
- View/download PDF
3. Non-CpG island promoter hypomethylation and miR-149 regulate the expression of SRPX2 in colorectal cancer
- Author
-
Øster B, Linnet L, Christensen LL, Thorsen K, Ongen H, Dermitzakis ET, Sandoval J, Moran S, Esteller M, Hansen TF, Lamy P, COLOFOL steering group, Laurberg S, Ørntoft TF, and Andersen CL
- Abstract
Gene silencing by DNA hypermethylation of CpG islands is a well-characterized phenomenon in cancer. The effect of hypomethylation in particular of non-CpG island genes is much less well described. By genome-wide screening, we identified 105 genes in microsatellite stable (MSS) colorectal adenocarcinomas with an inverse correlation (Spearman's ? = -0.40) between methylation and expression. Of these, 35 (33%) were hypomethylated non-CpG island genes and two of them, APOLD1 (Spearman's ? = -0.82) and SRPX2 (Spearman's ? = -0.80) were selected for further analyses. Hypomethylation of both genes were localized events not shared by adjacent genes. A set of 662 FFPE DNA samples not only confirmed that APOLD1 and SRPX2 are hypomethylated in CRC but also revealed hypomethylation to be significantly (p < 0.01) associated with tumors being localized in the left side, CpG island methylator phenotype negative, MSS, BRAF wt, undifferentiated and of adenocarcinoma histosubtype. Demethylation experiments supported SRPX2 being epigenetically regulated via DNA methylation, whereas other mechanisms in addition to DNA methylation seem to be involved in the regulation of APOLD1. We further identified miR-149 as a potential novel post-transcriptional regulator of SRPX2. In carcinoma tissue, miR-149 was downregulated and inversely correlated to SRPX2 (? = -0.77). Furthermore, ectopic expression of miR-149 significantly reduced SRPX2 transcript levels. Our study highlights that in colorectal tumors, hypomethylation of non-CpG island-associated promoters deregulate gene expression nearly as frequent as do CpG-island hypermethylation. The hypomethylation of SRPX2 is focal and not part of a large block. Furthermore, it often translates to an increased expression level, which may be modulated by miR-149.
- Published
- 2013
4. Maturation of dendritic cells by HHV-6B infection
- Author
-
Rasmussen, L., Petersen, Charlotte Christie, Øster, B., Höllsberg, Per, Agger, Ralf, and Hokland, M.E.
- Published
- 2006
5. Quantification of a human herpesvirus 6B-specific transcript by real-time PCR
- Author
-
Øster, B. and Höllsberg, P.
- Published
- 2002
6. Repression of KIAA1199 attenuates Wnt-signalling and decreases the proliferation of colon cancer cells.
- Author
-
Birkenkamp-Demtroder, K., Maghnouj, A., Mansilla, F., Thorsen, K., Andersen, C. L., Øster, B., Hahn, S., Ørntoft, T. F., Øster, B, and Ørntoft, T F
- Subjects
COLON cancer ,ADENOCARCINOMA ,RENAL cell carcinoma ,IMMUNOFLUORESCENCE ,FUNCTIONAL analysis - Abstract
Background: The KIAA1199 transcript is upregulated in colon adenomas and downregulated upon β-catenin knockdown.Methods: Transcript profiling was performed on >500 colon biopsies, methylation profiling data were compared with transcript data. Immunohistochemistry assessed KIAA1199 protein expression in 270 stage II/III tumours (>3 years follow-up). The effects of stable KIAA1199 knockdown in SW480 cells (three different constructs) were studied using transcriptional profiling, proliferation and protein analysis.Results: The KIAA1199 transcript was strongly upregulated in 95% of adenocarcinomas. Absent expression in normal mucosa correlated with KIAA1199 promotor methylation. Nuclear and cytoplasmic KIAA1199 protein expression was identified in colon adenocarcinomas and other types of cancers. A subpopulation of patients with tumours strongly expressing KIAA1199 in the nucleus showed a better outcome with regard to recurrence as lung or liver metastases. The KIAA1199 knockdown affected the cell cycle and the Wnt-signalling pathway. Reduced cellular proliferation and decreased KI67, phosphorylated retinoblastoma, β-catenin and ASCL2 protein expression supported these findings. Eighteen Wnt-signalling genes differentially expressed upon KIAA1199 knockdown correlated with the KIAA1199 expression profile in clinical specimens.Conclusion: The KIAA1199 knockdown attenuates the effects of the Wnt/β-catenin signalling and it may thus be regarded as a regulatory part of this pathway. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
7. Human herpesvirus 6B induces phosphorylation of p53 in its regulatory domain by a CK2- and p38-independent pathway
- Author
-
Øster, B., primary, Bundgaard, B., additional, Hupp, T. R., additional, and Höllsberg, P., additional
- Published
- 2008
- Full Text
- View/download PDF
8. Molecular-Subtype-Specific Biomarkers Improve Prediction of Prognosis in Colorectal Cancer.
- Author
-
Bramsen JB, Rasmussen MH, Ongen H, Mattesen TB, Ørntoft MW, Árnadóttir SS, Sandoval J, Laguna T, Vang S, Øster B, Lamy P, Madsen MR, Laurberg S, Esteller M, Dermitzakis ET, Ørntoft TF, and Andersen CL
- Subjects
- Biomarkers, Tumor genetics, Case-Control Studies, Colorectal Neoplasms classification, Colorectal Neoplasms pathology, Humans, Microbiota, Tumor Microenvironment, Biomarkers, Tumor classification, Colorectal Neoplasms genetics, Transcriptome
- Abstract
Colorectal cancer (CRC) is characterized by major inter-tumor diversity that complicates the prediction of disease and treatment outcomes. Recent efforts help resolve this by sub-classification of CRC into natural molecular subtypes; however, this strategy is not yet able to provide clinicians with improved tools for decision making. We here present an extended framework for CRC stratification that specifically aims to improve patient prognostication. Using transcriptional profiles from 1,100 CRCs, including >300 previously unpublished samples, we identify cancer cell and tumor archetypes and suggest the tumor microenvironment as a major prognostic determinant that can be influenced by the microbiome. Notably, our subtyping strategy allowed identification of archetype-specific prognostic biomarkers that provided information beyond and independent of UICC-TNM staging, MSI status, and consensus molecular subtyping. The results illustrate that our extended subtyping framework, combining subtyping and subtype-specific biomarkers, could contribute to improved patient prognostication and may form a strong basis for future studies., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
9. Functional screening identifies miRNAs influencing apoptosis and proliferation in colorectal cancer.
- Author
-
Christensen LL, Holm A, Rantala J, Kallioniemi O, Rasmussen MH, Ostenfeld MS, Dagnaes-Hansen F, Øster B, Schepeler T, Tobiasen H, Thorsen K, Sieber OM, Gibbs P, Lamy P, Hansen TF, Jakobsen A, Riising EM, Helin K, Lubinski J, Hagemann-Madsen R, Laurberg S, Ørntoft TF, and Andersen CL
- Subjects
- Aged, Aged, 80 and over, Animals, Cell Proliferation, DNA Methylation genetics, Down-Regulation genetics, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, HCT116 Cells, Humans, Intestinal Mucosa pathology, Laser Capture Microdissection, Male, Mice, MicroRNAs genetics, Middle Aged, Neoplasm Proteins metabolism, Neoplasm Staging, Phenotype, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Small Interfering metabolism, Reproducibility of Results, Apoptosis genetics, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, High-Throughput Screening Assays, MicroRNAs metabolism
- Abstract
MicroRNAs (miRNAs) play a critical role in many biological processes and are aberrantly expressed in human cancers. Particular miRNAs function either as tumor suppressors or oncogenes and appear to have diagnostic and prognostic significance. Although numerous miRNAs are dys-regulated in colorectal cancer (CRC) only a small fraction has been characterized functionally. Using high-throughput functional screening and miRNA profiling of clinical samples the present study aims at identifying miRNAs important for the control of cellular growth and/or apoptosis in CRC. The high-throughput functional screening was carried out in six CRC cell lines transfected with a pre-miR library including 319 synthetic human pre-miRs. Phenotypic alterations were evaluated by immunostaining of cleaved cPARP (apoptosis) or MKI67 (proliferation). Additionally, TaqMan Human MicroRNA Array Set v2.0 was used to profile the expression of 667 miRNAs in 14 normal colon mucosa and 46 microsatellite stable stage II CRC patients. Among the miRNAs that induced growth arrest and apoptosis in the CRC cell lines, and at same time were dys-regulated in the clinical samples, miR-375 was selected for further analysis. Independent in vitro analysis of transient and stable transfected CRC cell lines confirmed that miR-375 reduces cell viability through the induction of apoptotic death. We identified YAP1 as a direct miR-375 target in CRC and show that HELLS and NOLC1 are down-stream targets. Knock-down of YAP1 mimicked the phenotype induced by miR-375 over-expression indicating that miR-375 most likely exerts its pro-apoptotic role through YAP1 and its anti-apoptotic down-stream targets BIRC5 and BCL2L1. Finally, in vivo analysis of mouse xenograft tumors showed that miR-375 expression significantly reduced tumor growth. We conclude that the high-throughput screening successfully identified miRNAs that induce apoptosis and/or inhibit proliferation in CRC cells. Finally, combining the functional screening with profiling of CRC tissue samples we identified clinically relevant miRNAs and miRNA targets in CRC.
- Published
- 2014
- Full Text
- View/download PDF
10. Keratin23 (KRT23) knockdown decreases proliferation and affects the DNA damage response of colon cancer cells.
- Author
-
Birkenkamp-Demtröder K, Hahn SA, Mansilla F, Thorsen K, Maghnouj A, Christensen R, Øster B, and Ørntoft TF
- Subjects
- Base Sequence, Blotting, Western, Cell Cycle genetics, Cell Line, Tumor, Cell Survival genetics, Cell Survival radiation effects, Colonic Neoplasms genetics, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, DNA Damage, DNA Methylation, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Gamma Rays, Gene Expression Regulation, Neoplastic genetics, HCT116 Cells, HEK293 Cells, Humans, Keratins, Type I metabolism, MRE11 Homologue Protein, Microscopy, Fluorescence, Molecular Sequence Data, Oligonucleotide Array Sequence Analysis, Rad51 Recombinase genetics, Rad51 Recombinase metabolism, Reverse Transcriptase Polymerase Chain Reaction, Transcriptome genetics, Cell Proliferation, DNA Repair genetics, Keratins, Type I genetics, RNA Interference
- Abstract
Keratin 23 (KRT23) is strongly expressed in colon adenocarcinomas but absent in normal colon mucosa. Array based methylation profiling of 40 colon samples showed that the promoter of KRT23 was methylated in normal colon mucosa, while hypomethylated in most adenocarcinomas. Promoter methylation correlated with absent expression, while increased KRT23 expression in tumor samples correlated with promoter hypomethylation, as confirmed by bisulfite sequencing. Demethylation induced KRT23 expression in vitro. Expression profiling of shRNA mediated stable KRT23 knockdown in colon cancer cell lines showed that KRT23 depletion affected molecules of the cell cycle and DNA replication, recombination and repair. In vitro analyses confirmed that KRT23 depletion significantly decreased the cellular proliferation of SW948 and LS1034 cells and markedly decreased the expression of genes involved in DNA damage response, mainly molecules of the double strand break repair homologous recombination pathway. KRT23 knockdown decreased the transcript and protein expression of key molecules as e.g. MRE11A, E2F1, RAD51 and BRCA1. Knockdown of KRT23 rendered colon cancer cells more sensitive to irradiation and reduced proliferation of the KRT23 depleted cells compared to irradiated control cells.
- Published
- 2013
- Full Text
- View/download PDF
11. Inhibition of p53-dependent, but not p53-independent, cell death by U19 protein from human herpesvirus 6B.
- Author
-
Kofod-Olsen E, Møller JM, Schleimann MH, Bundgaard B, Bak RO, Øster B, Mikkelsen JG, Hupp T, and Höllsberg P
- Subjects
- Cell Death radiation effects, DNA Damage, Gamma Rays, Gene Expression Regulation radiation effects, HCT116 Cells, Herpesvirus 6, Human metabolism, Humans, Microarray Analysis, Signal Transduction radiation effects, Tumor Suppressor Protein p53 metabolism, Viral Proteins metabolism, Herpesvirus 6, Human genetics, Host-Pathogen Interactions, Tumor Suppressor Protein p53 genetics, Viral Proteins genetics
- Abstract
Infection with human herpesvirus (HHV)-6B alters cell cycle progression and stabilizes tumor suppressor protein p53. In this study, we have analyzed the activity of p53 after stimulation with p53-dependent and -independent DNA damaging agents during HHV-6B infection. Microarray analysis, Western blotting and confocal microscopy demonstrated that HHV-6B-infected cells were resistant to p53-dependent arrest and cell death after γ irradiation in both permissive and non-permissive cell lines. In contrast, HHV-6B-infected cells died normally through p53-independet DNA damage induced by UV radiation. Moreover, we identified a viral protein involved in inhibition of p53 during HHV-6B-infection. The protein product from the U19 ORF was able to inhibit p53-dependent signaling following γ irradiation in a manner similar to that observed during infection. Similar to HHV-6B infection, overexpression of U19 failed to rescue the cells from p53-independent death induced by UV radiation. Hence, infection with HHV-6B specifically blocks DNA damage-induced cell death associated with p53 without inhibiting the p53-independent cell death response. This block in p53 function can in part be ascribed to the activities of the viral U19 protein.
- Published
- 2013
- Full Text
- View/download PDF
12. Frequent genomic loss at chr16p13.2 is associated with poor prognosis in colorectal cancer.
- Author
-
Andersen CL, Lamy P, Thorsen K, Kjeldsen E, Wikman F, Villesen P, Øster B, Laurberg S, and Ørntoft TF
- Subjects
- Aged, Cell Line, Tumor, Disease-Free Survival, Female, Humans, Male, Oligonucleotide Array Sequence Analysis, Polymorphism, Single Nucleotide, Prognosis, Adenoma genetics, Carcinoma genetics, Chromosomes, Human, Pair 16, Colorectal Neoplasms genetics, DNA Copy Number Variations
- Abstract
Genomic alterations play important roles in colorectal cancer (CRC) carcinogenesis. Here, we aimed to identify and characterize recurrent copy-number alterations (CNAs) associated with clinical outcome of CRC by the use of single nucleotide polymorphism arrays, genomic quantitative PCR (qPCR) and fluorescence in situ hybridization (FISH). Colorectal neoplasia specimens and paired germline samples from 144 patients (40 adenomas and 104 carcinomas) as well as 40 CRC cell lines were investigated. This large dataset revealed frequent loss, including homozygous loss, at chr16p13.2 (from 5.9 to 7.42Mb). The loss was observed in 30% of adenomas and even more frequently in carcinomas, 56%, indicating that the loss define a subset of adenomas with a propensity for invasion. Consistent with this, the loss occurred twice as frequent in villous (40%) as in tubular adenomas (20%). The loss occurred independently of microsatellite stability and could be validated by qPCR in an independent sample cohort (n = 71). In Stage II/III, microsatellite stable (MSS) CRC it was associated with poor recurrence free survival (hazard ratio 2.4; p = 0.02; Multivariate Cox regression analysis). No transcriptional consequences of the losses were observed, and the only gene, A2BP1, located in the region showed no mutations. Correlation with other CNAs was established for chr3p22 in carcinomas and chr20p (inverse) in adenomas. FISH documented the chr16p13.2 region to be involved in complex structural rearrangements that included translocation to chr3p22 in some cases. The findings indicate that structural rearrangements involving chr16p13.2 are very frequent in colorectal neoplasia, often lead to homozygous deletion, and are associated with poor clinical outcome., (Copyright © 2011 UICC.)
- Published
- 2011
- Full Text
- View/download PDF
13. Tumor-specific usage of alternative transcription start sites in colorectal cancer identified by genome-wide exon array analysis.
- Author
-
Thorsen K, Schepeler T, Øster B, Rasmussen MH, Vang S, Wang K, Hansen KQ, Lamy P, Pedersen JS, Eller A, Mansilla F, Laurila K, Wiuf C, Laurberg S, Dyrskjøt L, Ørntoft TF, and Andersen CL
- Subjects
- Adaptor Proteins, Vesicular Transport genetics, Adaptor Proteins, Vesicular Transport metabolism, Alternative Splicing, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Carrier Proteins genetics, Carrier Proteins metabolism, Checkpoint Kinase 1, Cohort Studies, Colorectal Neoplasms pathology, Humans, Oligonucleotide Array Sequence Analysis, Promoter Regions, Genetic, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Kinases genetics, Protein Kinases metabolism, RNA, Messenger metabolism, Receptors, Steroid, Wnt Signaling Pathway, Oxysterol Binding Proteins, Colorectal Neoplasms genetics, Exons, Transcription Initiation Site
- Abstract
Background: Approximately half of all human genes use alternative transcription start sites (TSSs) to control mRNA levels and broaden the transcriptional output in healthy tissues. Aberrant expression patterns promoting carcinogenesis, however, may arise from alternative promoter usage., Results: By profiling 108 colorectal samples using exon arrays, we identified nine genes (TCF12, OSBPL1A, TRAK1, ANK3, CHEK1, UGP2, LMO7, ACSL5, and SCIN) showing tumor-specific alternative TSS usage in both adenoma and cancer samples relative to normal mucosa. Analysis of independent exon array data sets corroborated these findings. Additionally, we confirmed the observed patterns for selected mRNAs using quantitative real-time reverse-transcription PCR. Interestingly, for some of the genes, the tumor-specific TSS usage was not restricted to colorectal cancer. A comprehensive survey of the nine genes in lung, bladder, liver, prostate, gastric, and brain cancer revealed significantly altered mRNA isoform ratios for CHEK1, OSBPL1A, and TCF12 in a subset of these cancer types.To identify the mechanism responsible for the shift in alternative TSS usage, we antagonized the Wnt-signaling pathway in DLD1 and Ls174T colorectal cancer cell lines, which remarkably led to a shift in the preferred TSS for both OSBPL1A and TRAK1. This indicated a regulatory role of the Wnt pathway in selecting TSS, possibly also involving TP53 and SOX9, as their transcription binding sites were enriched in the promoters of the tumor preferred isoforms together with their mRNA levels being increased in tumor samples. Finally, to evaluate the prognostic impact of the altered TSS usage, immunohistochemistry was used to show deregulation of the total protein levels of both TCF12 and OSBPL1A, corresponding to the mRNA levels observed. Furthermore, the level of nuclear TCF12 had a significant correlation to progression free survival in a cohort of 248 stage II colorectal cancer samples., Conclusions: Alternative TSS usage in colorectal adenoma and cancer samples has been shown for nine genes, and OSBPL1A and TRAK1 were found to be regulated in vitro by Wnt signaling. TCF12 protein expression was upregulated in cancer samples and correlated with progression free survival.
- Published
- 2011
- Full Text
- View/download PDF
14. Alternative splicing of SLC39A14 in colorectal cancer is regulated by the Wnt pathway.
- Author
-
Thorsen K, Mansilla F, Schepeler T, Øster B, Rasmussen MH, Dyrskjøt L, Karni R, Akerman M, Krainer AR, Laurberg S, Andersen CL, and Ørntoft TF
- Subjects
- Base Sequence, Binding Sites, Cation Transport Proteins metabolism, Cell Line, Tumor, Colorectal Neoplasms enzymology, Exons genetics, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Humans, Intestinal Mucosa enzymology, Intestinal Mucosa pathology, Introns genetics, Nuclear Proteins genetics, Nuclear Proteins metabolism, Protein Binding, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Serine-Arginine Splicing Factors, Alternative Splicing genetics, Cation Transport Proteins genetics, Colorectal Neoplasms genetics, Signal Transduction, Wnt Proteins metabolism
- Abstract
Alternative splicing is a crucial step in the generation of protein diversity and its misregulation is observed in many human cancer types. By analyzing 143 colorectal samples using exon arrays, SLC39A14, a divalent cation transporter, was identified as being aberrantly spliced in tumor samples. SLC39A14 contains two mutually exclusive exons 4A and 4B and the exon 4A/4B ratio was significantly altered in adenomas (p = 3.6 × 10(-10)) and cancers (p = 9.4 × 10(-11)), independent of microsatellite stability status. The findings were validated in independent exon array data sets and by quantitative real-time reverse-transcription PCR (qRT-PCR). Aberrant Wnt signaling is a hallmark of colorectal tumorigenesis and is characterized by nuclear β-catenin. Experimental inactivation of Wnt signaling in DLD1 and Ls174T cells by knockdown of β-catenin or overexpression of dominant negative TCFs (TCF1 and TCF4) altered the 4A/4B ratio, indicating that SLC39A14 splicing is regulated by the Wnt pathway. An altered 4A/4B ratio was also observed in gastric and lung cancer where Wnt signaling is also known to be aberrantly activated. The splicing factor SRSF1 and its regulator, the kinase SRPK1, were found to be deregulated upon Wnt inactivation in colorectal carcinoma cells. SRPK1 was also found up-regulated in both adenoma samples (p = 1.5 × 10(-5)) and cancer samples (p = 5 × 10(-4)). In silico splicing factor binding analysis predicted SRSF1 to bind predominantly to the cancer associated exon 4B, hence, it was hypothesized that SRPK1 activates SRSF1 through phosphorylation, followed by SRSF1 binding to exon 4B and regulation of SLC39A14 splicing. Indeed, siRNA-mediated knockdown of SRPK1 and SRSF1 in DLD1 and SW480 colorectal cancer cells led to a change in the 4A/4B isoform ratio, supporting a role of these factors in the regulation of SLC39A14 splicing. In conclusion, alternative splicing of SLC39A14 was identified in colorectal tumors and found to be regulated by the Wnt pathway, most likely through regulation of SRPK1 and SRSF1.
- Published
- 2011
- Full Text
- View/download PDF
15. Restriction of human herpesvirus 6B replication by p53.
- Author
-
Øster B, Kofod-Olsen E, Bundgaard B, and Höllsberg P
- Subjects
- Benzothiazoles pharmacology, Cell Line, Cytopathogenic Effect, Viral, Enzyme Inhibitors pharmacology, Epithelial Cells virology, Humans, Toluene analogs & derivatives, Toluene pharmacology, Tumor Suppressor Protein p53 antagonists & inhibitors, Tumor Suppressor Protein p53 deficiency, Herpesvirus 6, Human immunology, Tumor Suppressor Protein p53 physiology, Virus Replication immunology
- Abstract
Human herpesvirus 6B (HHV-6B) induces significant accumulation of p53 in both the nucleus and cytoplasm during infection. Activation of p53 by DNA damage is known to induce either growth arrest or apoptosis; nevertheless, HHV-6B-infected cells are arrested in their cell cycle independently of p53, and only a minor fraction of the infected cells undergoes apoptosis. Using pifithrin-alpha, a p53 inhibitor, and p53-null cells, this study showed that infected epithelial cells accumulated viral transcripts and proteins to a significantly higher degree in the absence of active p53. Moreover, HHV-6B-induced cytopathic effects were greatly enhanced in the absence of p53. This suggests that, in epithelial cells, some of the functions of p53 leading to cell-cycle arrest and apoptosis are restrained by HHV-6B infection, whereas other cellular defences, causing inhibition of virus transcription, are partially retained.
- Published
- 2008
- Full Text
- View/download PDF
16. Human herpesvirus 6B inhibits cell proliferation by a p53-independent pathway.
- Author
-
Øster B, Kaspersen MD, Kofod-Olsen E, Bundgaard B, and Höllsberg P
- Subjects
- Benzothiazoles pharmacology, Cell Cycle, Cell Line, Cyclin-Dependent Kinase Inhibitor p21 biosynthesis, DNA-Binding Proteins biosynthesis, Humans, Toluene analogs & derivatives, Toluene pharmacology, Tumor Suppressor Protein p53 antagonists & inhibitors, Tumor Suppressor Protein p53 biosynthesis, Viral Proteins biosynthesis, Virus Replication, Cell Proliferation, Herpesvirus 6, Human physiology, Tumor Suppressor Protein p53 physiology
- Abstract
Background: Various forms of cellular stress can activate the tumour suppressor protein p53, an important regulator of cell cycle arrest, apoptosis, and cellular senescence. Cells infected by human herpesvirus 6B (HHV-6B) accumulate aberrant amounts of p53., Objectives: The aim of this study was to investigate the role of p53 accumulation in the HHV-6B-induced cell cycle arrest., Study Design: The role of p53 was studied using the p53 inhibitor pifithrin-a, and cells genetically deficient in functional p53 by homologous recombination., Results: In response to HHV-6B infection, epithelial cells were arrested in the G1/S phase of the cell cycle concomitant with an aberrant accumulation of p53. However, the known p53-induced mediator of cell cycle arrest, p21, was not upregulated. Approximately 90% of the cells expressed HHV-6B p41, indicative of viral infection. The presence of pifithrin-a, a p53 inhibitor, did not reverse the HHV-6B-induced cell cycle block. In support of this, HHV-6B infection of p53(-/-) cells induced a cell cycle block before S-phase with kinetics similar to or faster than that observed by infection in wt cells., Conclusions: HHV-6B infection inhibited host cell proliferation concomitantly with p53 accumulation, but importantly the block in cell cycle occurred by a pathway independent of p53.
- Published
- 2006
- Full Text
- View/download PDF
17. Human herpesvirus 6B induces cell cycle arrest concomitant with p53 phosphorylation and accumulation in T cells.
- Author
-
Øster B, Bundgaard B, and Höllsberg P
- Subjects
- Cell Line, DNA-Binding Proteins metabolism, Humans, Phosphorylation, Roseolovirus Infections virology, T-Lymphocytes cytology, T-Lymphocytes metabolism, Cell Cycle physiology, Herpesvirus 6, Human pathogenicity, T-Lymphocytes virology, Tumor Suppressor Protein p53 metabolism
- Abstract
We studied the interactions between human herpesvirus 6B (HHV-6B) and its host cell. Productive infections of T-cell lines led to G1/S- and G2/M-phase arrest in the cell cycle concomitant with an increased level and enhanced DNA-binding activity of p53. More than 70% of HHV-6B-infected cells did not bind annexin V, indicating that the majority of cells were not undergoing apoptosis. HHV-6B infection induced Ser20 and Ser15 phosphorylation on p53, and the latter was inhibited by caffeine, an ataxia telangiectasia mutated kinase inhibitor. Thus, a productive HHV-6B infection suppresses T-cell proliferation concomitant with the phosphorylation and accumulation of p53.
- Published
- 2005
- Full Text
- View/download PDF
18. A Sensitive Quantification of HHV-6B by Real-time PCR.
- Author
-
ØSter B and Höllsberg P
- Abstract
Human herpesvirus (HHV)-6B is a pathogen causing latent infection in virtually all humans. Nevertheless, the interaction of HHV-6B with its host cells is poorly understood. Although HHV-6B is approximately 90% homologous to HHV-6A, it expresses certain B-specific genes. In order to quantify the amount of expressed viral mRNA we have developed a method using real-time PCR on a LightCycler instrument. Here we describe an assay for the detection of the HHV-6B B6 mRNA, but our approach can easily be extended to involve other mRNAs. This method is useful during the study of HHV-6B biology and offers reliable and reproducible, quantitative detection of viral mRNA below the attomol range.
- Published
- 2002
- Full Text
- View/download PDF
19. Viral gene expression patterns in human herpesvirus 6B-infected T cells.
- Author
-
Øster B and Höllsberg P
- Subjects
- Base Sequence, Cell Line, DNA-Directed DNA Polymerase metabolism, Herpesvirus 6, Human genetics, Humans, Immediate-Early Proteins, Molecular Sequence Data, Transcription, Genetic, Viral Proteins metabolism, Gene Expression Regulation, Viral, Herpesvirus 6, Human metabolism, T-Lymphocytes virology, Viral Proteins genetics
- Abstract
Herpesvirus gene expression is divided into immediate-early (IE) or alpha genes, early (E) or beta genes, and late (L) or gamma genes on the basis of temporal expression and dependency on other gene products. By using real-time PCR, we have investigated the expression of 35 human herpesvirus 6B (HHV-6B) genes in T cells infected by strain PL-1. Kinetic analysis and dependency on de novo protein synthesis and viral DNA polymerase activity suggest that the HHV-6B genes segregate into six separate kinetic groups. The genes expressed early (groups I and II) and late (groups V and VI) corresponded well with IE and L genes, whereas the intermediate groups III and IV contained E and L genes. Although HHV-6B has characteristics similar to those of other roseoloviruses in its overall gene regulation, we detected three B-variant-specific IE genes. Moreover, genes that were independent of de novo protein synthesis clustered in an area of the viral genome that has the lowest identity to the HHV-6A variant. The organization of IE genes in an area of the genome that differs from that of HHV-6A underscores the distinct differences between HHV-6B and HHV-6A and may provide a basis for further molecular and immunological analyses to elucidate their different biological behaviors.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.