Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Celina Cziepluch is active.

Publication


Featured researches published by Celina Cziepluch.


The EMBO Journal | 1991

The N-Myc oncoprotein is associated in vivo with the phosphoprotein Max(p20/22) in human neuroblastoma cells.

Achim Wenzel; Celina Cziepluch; Ute Hamann; Jörg Schürmann; Manfred Schwab

Proteins encoded by the proto‐oncogenes c‐myc, L‐myc, and N‐myc contain at their carboxy‐terminus a tripartite segment comprising a basic DNA binding region (BR), a helix‐loop‐helix (HLH) and a leucine zipper motif (Zip), that are believed to be involved in DNA binding and protein‐protein interaction. The N‐Myc oncoprotein is overexpressed in certain human tumors that share neuroectodermal features due to amplification of the N‐myc gene. Using a monoclonal antibody directed against an N‐terminal epitope of the N‐Myc protein in immunoprecipitations performed with extracts of neuroblastoma cells, two nuclear phosphoprotein, p20/22, forming a hetero‐oligomeric complex with N‐Myc are identified. Both proteins are phosphorylated by casein kinase II in vitro. By partial proteolytic maps we show that p20 and p22 are structurally related to each other and that p20 is identical with Max, a recently described in vitro binding partner of myc proteins. Time course experiments show the presence of the complex in cellular extracts immunoprecipitated within a 5 min interval after the preparation of the cell extract. While the expression of N‐myc is restricted, expression of both Max(p20/22) and the murine homolog Myn(p20/22) was observed in cells of diverse human and murine embryonal lineages as detected by heterologous complex formation. By introduction of expression vectors containing the wild type N‐myc gene or N‐myc genes with in frame deletions or point mutations into recipient cells and subsequent immunoprecipitation of the resulting N‐Myc proteins we show that the HLH‐Zip region is essential to the formation of the N‐Myc‐p20/22 complex.


Journal of Virology | 2001

In Vivo Accumulation of Cyclin A and Cellular Replication Factors in Autonomous Parvovirus Minute Virus of Mice-Associated Replication Bodies

Tarig Bashir; Jean Rommelaere; Celina Cziepluch

ABSTRACT Autonomous parvovirus minute virus of mice (MVM) DNA replication is strictly dependent on cellular factors expressed during the S phase of the cell cycle. Here we report that MVM DNA replication proceeds in specific nuclear structures termed autonomous parvovirus-associated replication bodies, where components of the basic cellular replication machinery accumulate. The presence of DNA polymerases α and δ in these bodies suggests that MVM utilizes partially preformed cellular replication complexes for its replication. The recruitment of cyclin A points to a role for this cell cycle factor in MVM DNA replication beyond its involvement in activating the conversion of virion single-stranded DNA to the duplex replicative form.


Journal of Virology | 2000

H-1 Parvovirus-Associated Replication Bodies: a Distinct Virus-Induced Nuclear Structure

Celina Cziepluch; Stefan Lampel; Annabel Grewenig; Christine Grund; Peter Lichter; Jean Rommelaere

We have identified a nuclear structure that is induced after infection with the autonomous parvovirus H-1. Using fluorescence microscopy, we observed that the major nonstructural protein (NS1) of H-1 virus which is essential for viral DNA amplification colocalized with virus-specific DNA sequences and sites of ongoing viral DNA replication in distinct nuclear bodies which we designated H-1 parvovirus-associated replication bodies (H-1 PAR-bodies). In addition, two cellular proteins were shown to accumulate in H1 PAR-bodies: (i) the proliferating cell nuclear antigen (PCNA) which is essential for chromosomal and parvoviral replication and (ii) the NS1-interacting small glutamine-rich TPR-containing protein (SGT), suggesting a role for the latter in parvoviral replication and/or gene expression. Since many DNA viruses target preexisting nuclear structures, known as PML-bodies, for viral replication and gene expression, we have determined the localization of H-1 PAR- and PML-bodies by double-fluorescence labeling and confocal microscopy and found them to be spatially unrelated. Furthermore, H-1 PAR-bodies did not colocalize with other prominent nuclear structures such as nucleoli, coiled bodies, and speckled domains. Electron microscopy analysis revealed that NS1, as detected by indirect immunogold labeling, was localized in ring-shaped electron-dense nuclear structures corresponding in size and frequency to H-1 PAR-bodies. These structures were also clearly visible without immunogold labeling and could be detected only in infected cells. Our results suggest that H-1 virus does not target known nuclear bodies for DNA replication but rather induces the formation of a novel structure in the nucleus of infected cells.


Journal of Virology | 2014

Complementary induction of immunogenic cell death by oncolytic parvovirus H-1PV and gemcitabine in pancreatic cancer

Assia L. Angelova; Svitlana P. Grekova; Anette Heller; Olga Kuhlmann; Esther Soyka; Thomas Giese; Marc Aprahamian; Gaétan Bour; Sven Rüffer; Celina Cziepluch; Laurent Daeffler; Jean Rommelaere; Jens Werner; Zahari Raykov; Nathalia A. Giese

ABSTRACT Novel therapies employing oncolytic viruses have emerged as promising anticancer modalities. The cure of particularly aggressive malignancies requires induction of immunogenic cell death (ICD), coupling oncolysis with immune responses via calreticulin, ATP, and high-mobility group box protein B1 (HMGB1) release from dying tumor cells. The present study shows that in human pancreatic cancer cells (pancreatic ductal adenocarcinoma [PDAC] cells; n = 4), oncolytic parvovirus H-1 (H-1PV) activated multiple interconnected death pathways but failed to induce calreticulin exposure or ATP release. In contrast, H-1PV elevated extracellular HMGB1 levels by 4.0 ± 0.5 times (58% ± 9% of total content; up to 100 ng/ml) in all infected cultures, whether nondying, necrotic, or apoptotic. An alternative secretory route allowed H-1PV to overcome the failure of gemcitabine to trigger HMGB1 release, without impeding cytotoxicity or other ICD activities of the standard PDAC medication. Such broad resistance of H-1PV-induced HMGB1 release to apoptotic blockage coincided with but was uncoupled from an autocrine interleukin-1β (IL-1β) loop. That and the pattern of viral determinants maintained in gemcitabine-treated cells suggested the activation of an inflammasome/caspase 1 (CASP1) platform alongside DNA detachment and/or nuclear exclusion of HMGB1 during early stages of the viral life cycle. We concluded that H-1PV infection of PDAC cells is signaled through secretion of the alarmin HMGB1 and, besides its own oncolytic effect, might convert drug-induced apoptosis into an ICD process. A transient arrest of cells in the cyclin A1-rich S phase would suffice to support compatibility of proliferation-dependent H-1PV with cytotoxic regimens. These properties warrant incorporation of the oncolytic virus H-1PV, which is not pathogenic in humans, into multimodal anticancer treatments. IMPORTANCE The current therapeutic concepts targeting aggressive malignancies require an induction of immunogenic cell death characterized by exposure of calreticulin (CRT) as well as release of ATP and HMGB1 from dying cells. In pancreatic tumor cells (PDAC cells) infected with the oncolytic parvovirus H-1PV, only HMGB1 was released by all infected cells, whether nondying, necrotic, or succumbing to one of the programmed death pathways, including contraproductive apoptosis. Our data suggest that active secretion of HMGB1 from PDAC cells is a sentinel reaction emerging during early stages of the viral life cycle, irrespective of cell death, that is compatible with and complements cytotoxic regimens. Consistent induction of HMGB1 secretion raised the possibility that this reaction might be a general “alarming” phenomenon characteristic of H-1PVs interaction with the host cell; release of IL-1β points to the possible involvement of a danger-sensing inflammasome platform. Both provide a basis for further virus-oriented studies.


Cancer Biology & Therapy | 2010

Immune cells participate in the oncosuppressive activity of parvovirus H-1PV and are activated as a result of their abortive infection with this agent.

Svitlana P. Grekova; Marc Aprahamian; Nathalia A. Giese; Steffen Schmitt; Thomas Giese; Christine S. Falk; Laurent Daeffler; Celina Cziepluch; Jean Rommelaere; Zahari Raykov

Treatment of cancers by means of viruses, that specifically replicate in (oncotropism) and kill (oncolysis) neoplastic cells, is increasingly gaining acceptance in the clinic. Among these agents, parvoviruses have been shown to possess not only direct oncolytic but also immunomodulating properties, serving as an adjuvant to prime the immune system to react against infected tumors. Here, we aimed to establish whether immunomodulating mechanisms participate in the recently reported therapeutic potential of parvoviruses against pancreatic carcinoma. Using adoptive transfer experiments we discovered that the transfer of splenocytes of donor rats harboring H-1PV-treated orthotopic PDAC tumors could significantly prolong the survival of naïve tumor-bearing recipients, compared to those receiving cells from mock-treated donors. Closer investigation of immunological parameters in infected donor rats revealed that virus-induced interferon gamma production and cellular immune response played an important role in this effect. These data have also preclinical relevance since abortive H-1PV infection of human peripheral blood mononuclear cells or cocultivation of these cells with H-1PV-preinfected pancreatic cancer cells, resulted in enhancement of innate and adaptive immune reactivity. Taken together our data reveal that oncolytic H-1PV modulates the immune system into an anticancer state, and further support the concept of using parvoviruses in the fight against pancreatic cancer.


Journal of Virology | 2010

Activation of an Antiviral Response in Normal but Not Transformed Mouse Cells: a New Determinant of Minute Virus of Mice Oncotropism

Svitlana P. Grekova; Rainer Zawatzky; Rita Hörlein; Celina Cziepluch; Michal Mincberg; Claytus Davis; Jean Rommelaere; Laurent Daeffler

ABSTRACT Parvovirus minute virus of mice (MVMp) is endowed with oncotropic properties so far ascribed only to the dependency of the virus life cycle on cellular factors expressed during S phase and/or modulated by malignant transformation. For other viruses oncotropism relies on their inability to circumvent type I interferon (IFN)-induced innate antiviral mechanisms, the first line of defense triggered by normal cells against viral infections. These agents propagate, therefore, preferentially in transformed/tumor cells, which often lack functional antiviral mechanisms. The present study aimed at investigating whether antiviral processes also contribute to MVMp oncotropism. Our results demonstrate that in contrast to MVMp-permissive transformed mouse A9 fibroblasts, freshly isolated normal counterparts (mouse embryonic fibroblasts [MEFs]) mount, through production and release of type I IFNs upon their infection, an antiviral response against MVMp lytic multiplication. Pretreatment of MEFs with a type I IFN-β-neutralizing antibody, prior to MVMp infection, inhibits the virus-triggered antiviral response and improves the fulfillment of the MVMp life cycle. Our results also show that part of the A9 permissiveness to MVMp relies on the inability to produce type I IFNs upon parvovirus infection, a feature related either to an A9 intrinsic deficiency of this process or to an MVMp-triggered inhibitory mechanism, since stimulation of these cells by exogenous IFN-β strongly inhibits the parvovirus life cycle. Taken together, our results demonstrate for the first time that parvovirus infection triggers an innate antiviral response in normal cells and suggest that the MVMp oncotropism depends at least in part on the failure of infected transformed cells to mount such a response.


Yeast | 1997

Sequence and Analysis of a 36·2 kb Fragment from the Right Arm of Yeast Chromosome XV Reveals 19 Open Reading Frames Including SNF2 (5′ end), CPA1, SLY41, a Putative Transport ATPase, a Putative Ribosomal Protein and an SNF2 Homologue

Rémy Poirey; Celina Cziepluch; Edda Tobiasch; Aurora Pujol; Elisabeth Kordes; Jean Claude Jauniaux

The complete sequence of a 36 196 bp DNA segment located on the right arm of chromosome XV of Saccharomyces cerevisiae has been determined and analysed. The sequence includes the 5′ coding region of the SNF2 gene, the CPA1 leader peptide sequence and 17 open reading frames (ORFs) of at least 100 amino acids. Two of these correspond to previously known genes (CPA1, SLY41), whereas 15 correspond to new genes. The putative translation products of three ORFs show significant similarity with known proteins: one is a putative transport ATPase, another appears to be a ribosomal protein, and the third is an Snf2p homologue. The sequence has been deposited in the EMBL databank under Accession Numbers: Z75198, Z75199, Z75200, Z75201, Z75202, Z75203, Z75204, Z75205, Z75206, Z75207, Z75209, Z75210, Z75211, Z75212, Z75213, Z75214, Z75215, Z75216.


Archive | 1990

Genetic Alterations in Colorectal Cancers in Correlation to Clinical Parameters

Manfred Schwab; Silke Brüderlein; Kai Van Der Bosch; Andreas Weith; Ilona Leister; Celina Cziepluch; Lukas C. Amler; Peter M. Schlag

Genetic alterations resulting in oncogenic activation of certain cellular genes or in deletion of tumor suppressor genes may play an important role in colorectal cancers. Cytogenetic and molecular genetic approaches led to the identification of non-random genetic alterations including mutational activation of RAS genes, and deletion of genetic material from various chromosomal loci. Our own systematic study of the chromosomal status of colorectal cancer cells has more recently uncovered cytogenetic evidence for DNA amplification. Our data raise the suspicion that amplification is mainly associated with an advanced or metastatic tumor stage (Dukes C or D). The study of other cancers had made evident that DNA amplification is a useful predictor for clinical outcome. Once the molecular identity of the amplified DNA in colorectal cancer is defined it should be possible to find out if amplified molecular probes may be useful for predicting the clinical behaviour of colorectal cancers.


Genes, Chromosomes and Cancer | 1989

Neuroblastoma consensus deletion maps to 1p36.1–2

Andreas Weith; Tommy Martinsson; Celina Cziepluch; Silke Brüderlein; Lukas C. Amler; Frank Berthold; Manfred Schwab


Cancer Research | 1990

Human Colorectal Cancer: High Frequency of Deletions at Chromosome 1p35

Ilona Leister; Andreas Weith; Silke Brüderlein; Celina Cziepluch; Daoroong Kangwanpong; Peter M. Schlag; Manfred Schwab

Collaboration


Dive into the Celina Cziepluch's collaboration.

Top Co-Authors

Avatar

Manfred Schwab

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jean Claude Jauniaux

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Andreas Weith

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Laurent Daeffler

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Nathalia A. Giese

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Svitlana P. Grekova

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Achim Wenzel

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jörg Schürmann

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zahari Raykov

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge