Network


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

Hotspot


Dive into the research topics where Nicole Bäumer is active.

Publication


Featured researches published by Nicole Bäumer.


Molecular and Cellular Biology | 2004

The cyclin A1-CDK2 complex regulates DNA double-strand break repair.

Carsten Müller-Tidow; Ping Ji; Sven Diederichs; Jenny Potratz; Nicole Bäumer; Gabriele Köhler; Thomas Cauvet; Chunaram Choudary; Tiffany van der Meer; Wan Yu Iris Chan; Conrad Nieduszynski; William H. Colledge; Mark Carrington; H. Phillip Koeffler; Anja Restle; Lisa Wiesmüller; Joëlle Sobczak-Thépot; Wolfgang E. Berdel; Hubert Serve

ABSTRACT Vertebrates express two A-type cyclins; both associate with and activate the CDK2 protein kinase. Cyclin A1 is required in the male germ line, but its molecular functions are incompletely understood. We observed specific induction of cyclin A1 expression and promoter activity after UV and γ-irradiation which was mediated by p53. cyclin A1−/− cells showed increased radiosensitivity. To unravel a potential role of cyclin A1 in DNA repair, we performed a yeast triple hybrid screen and identified the Ku70 DNA repair protein as a binding partner and substrate of the cyclin A1-CDK2 complex. DNA double-strand break (DSB) repair was deficient in cyclin A1−/− cells. Further experiments indicated that A-type cyclins activate DNA DSB repair by mechanisms that depend on CDK2 activity and Ku proteins. Both cyclin A1 and cyclin A2 enhanced DSB repair by homologous recombination, but only cyclin A1 significantly activated nonhomologous end joining. DNA DSB repair was specific for A-type cyclins because cyclin E was ineffective. These findings establish a novel function for cyclin A1 and CDK2 in DNA DSB repair following radiation damage.


Blood | 2010

BCR-ABL enhances differentiation of long-term repopulating hematopoietic stem cells

Mirle Schemionek; Christian Elling; Ulrich Steidl; Nicole Bäumer; Ashley Hamilton; Tilmann Spieker; Joachim R. Göthert; Martin Stehling; Amy J. Wagers; Claudia S. Huettner; Daniel G. Tenen; Lara Tickenbrock; Wolfgang E. Berdel; Hubert Serve; Tessa L. Holyoake; Carsten Müller-Tidow; Steffen Koschmieder

In a previously developed inducible transgenic mouse model of chronic myeloid leukemia, we now demonstrate that the disease is transplantable using BCR-ABL(+) Lin(-)Sca-1(+)c-kit(+) (LSK) cells. Interestingly, the phenotype is more severe when unfractionated bone marrow cells are transplanted, yet neither progenitor cells (Lin(-)Sca-1(-)c-kit(+)), nor mature granulocytes (CD11b(+)Gr-1(+)), nor potential stem cell niche cells (CD45(-)Ter119(-)) are able to transmit the disease or alter the phenotype. The phenotype is largely independent of BCR-ABL priming before transplantation. However, prolonged BCR-ABL expression abrogates the potential of LSK cells to induce full-blown disease in secondary recipients and increases the fraction of multipotent progenitor cells at the expense of long-term hematopoietic stem cells (LT-HSCs) in the bone marrow. BCR-ABL alters the expression of genes involved in proliferation, survival, and hematopoietic development, probably contributing to the reduced LT-HSC frequency within BCR-ABL(+) LSK cells. Reversion of BCR-ABL, or treatment with imatinib, eradicates mature cells, whereas leukemic stem cells persist, giving rise to relapsed chronic myeloid leukemia on reinduction of BCR-ABL, or imatinib withdrawal. Our results suggest that BCR-ABL induces differentiation of LT-HSCs and decreases their self-renewal capacity.


Blood | 2012

Genome wide analysis of histone H3 acetylation patterns in AML identifies PRDX2 as an epigenetically silenced tumor suppressor gene

Shuchi Agrawal-Singh; Fabienne Isken; Konstantin Agelopoulos; Hans-Ulrich Klein; Nils H. Thoennissen; Gabriele Koehler; Antje Hascher; Nicole Bäumer; Wolfgang E. Berdel; Christian Thiede; Gerhard Ehninger; Anke Becker; Peter Schlenke; Yipeng Wang; Michael McClelland; Utz Krug; Steffen Koschmieder; Thomas Büchner; Dae-Yeul Yu; Shailendra Vikram Singh; Klaus Hansen; Hubert Serve; Martin Dugas; Carsten Müller-Tidow

With the use of ChIP on microarray assays in primary leukemia samples, we report that acute myeloid leukemia (AML) blasts exhibit significant alterations in histone H3 acetylation (H3Ac) levels at > 1000 genomic loci compared with CD34(+) progenitor cells. Importantly, core promoter regions tended to have lower H3Ac levels in AML compared with progenitor cells, which suggested that a large number of genes are epigenetically silenced in AML. Intriguingly, we identified peroxiredoxin 2 (PRDX2) as a novel potential tumor suppressor gene in AML. H3Ac was decreased at the PRDX2 gene promoter in AML, which correlated with low mRNA and protein expression. We also observed DNA hypermethylation at the PRDX2 promoter in AML. Low protein expression of the antioxidant PRDX2 gene was clinically associated with poor prognosis in patients with AML. Functionally, PRDX2 acted as inhibitor of myeloid cell growth by reducing levels of reactive oxygen species (ROS) generated in response to cytokines. Forced PRDX2 expression inhibited c-Myc-induced leukemogenesis in vivo on BM transplantation in mice. Taken together, epigenome-wide analyses of H3Ac in AML led to the identification of PRDX2 as an epigenetically silenced growth suppressor, suggesting a possible role of ROS in the malignant phenotype in AML.


Oncogene | 2005

Cyclin A1, the alternative A-type cyclin, contributes to G1/S cell cycle progression in somatic cells

Ping Ji; Shuchi Agrawal; Sven Diederichs; Nicole Bäumer; Annette Becker; Thomas Cauvet; Sascha Kowski; Carmela Beger; Karl Welte; Wolfgang E. Berdel; Hubert Serve; Carsten Müller-Tidow

Cyclin A1 is an alternative A-type cyclin that is essential for spermatogenesis, but it is also expressed in hematopoietic progenitor cells and in acute myeloid leukemia. Its functions during cell cycle progression of somatic cells are incompletely understood. Here, we have analysed the cell cycle functions of cyclin A1 in transformed and nontransformed cells. Murine embryonic fibroblasts derived from cyclin A1-deficient mice were significantly impaired in their proliferative capacity. In accordance, cyclin A1−/− cells accumulated in G1 and G2/M phase while the percentage of S phase cells decreased. Also, lectin stimulated splenic lymphocytes from cyclin A1−/− mice proliferated slower than their wild-type counterparts. Forced cyclin A1 overexpression in NIH3T3 cells and in U937 leukemic cells either by transient transfection or by retroviral infection enhanced S phase entry. Consequently, siRNA mediated silencing of cyclin A1 in highly cyclin A1 expressing ML1 leukemic cells significantly slowed S phase entry, decreased proliferation and inhibited colony formation. Taken together, these analyses demonstrate that cyclin A1 contributes to G1 to S cell cycle progression in somatic cells. Cyclin A1 overexpression enhances S phase entry consistent with an oncogenic function. Finally, cyclin A1 might be a therapeutic target since its silencing inhibited leukemia cell growth.


Clinical Cancer Research | 2015

Antibody-mediated delivery of anti-KRAS-siRNA in vivo overcomes therapy resistance in colon cancer

Sebastian Bäumer; Nicole Bäumer; Neele Appel; Lisa Terheyden; Julia Fremerey; Sonja Schelhaas; Eva Wardelmann; Frank Buchholz; Wolfgang E. Berdel; Carsten Müller-Tidow

Purpose: KRAS mutations are frequent driver mutations in multiple cancers. KRAS mutations also induce anti-EGFR antibody resistance in adenocarcinoma such as colon cancer. The aim of this study was to overcome anti-EGFR antibody resistance by coupling the antibody to KRAS-specific siRNA. Experimental Design: The anti-EGFR antibody was chemically coupled to siRNA. The resulting complex was tested for antibody binding efficiency, serum stability and ability to deliver siRNA to EGFR-expressing cells. Western blotting, viability, apoptosis, and colony formation assays were performed for efficacy evaluation in vitro. Furthermore, therapeutic activity of the antibody–KRAS-siRNA complexes was examined in in vivo xenograft mouse tumor models. Results: Antibody–siRNA complexes were targeted and internalized via the EGFR receptor. Upon internalization, target gene expression was strongly and specifically repressed, followed by a reduced proliferation and viability, and induced apoptosis of the cells in vitro. Clonogenic growth of mutant KRAS-bearing cells was suppressed by KRAS-siRNA–anti-EGFR antibody complexes. In xenograft mouse models, anti-EGFR antibody–KRAS-siRNA complexes significantly slowed tumor growth in anti-EGFR–resistant cells. Conclusions: The coupling of siRNA against KRAS to anti-EGFR antibodies provides a novel therapy approach for KRAS-mutated EGFR-positive cancer cells in vitro and in vivo. These findings provide an innovative approach for cancer-specific siRNA application and for enhanced therapeutic potential of monoclonal antibody therapy and personalized treatment of cancer entities. Clin Cancer Res; 21(6); 1383–94. ©2015 AACR.


Leukemia | 2008

Pim2 complements Flt3 wild-type receptor in hematopoietic progenitor cell transformation

Shuchi Agrawal; Steffen Koschmieder; Nicole Bäumer; N G P Reddy; Wolfgang E. Berdel; Carsten Müller-Tidow; Hubert Serve

Pim2 is a serine/threonine kinase expressed at high levels in several malignancies including acute leukemia. Pim2 protein is induced by oncogenic Fms-like tyrosine kinase-3 (Flt3)-internal tandem duplications (ITD), but not by Flt3 wild-type receptor (Flt3-Wt) in response to Flt3 ligand (FL). Here we show that Pim2 can complement Flt3-Wt signaling and induce transformation similar to Flt3-ITD in myeloid cells. Our data demonstrate that Pim2 is necessary but not sufficient for Flt3-ITD-induced transformation of 32D cells and primary bone marrow cells as assessed by colony assays. Pim2-induced clonogenic growth of FL-treated 32D-Flt3-Wt cells. Proliferation of 32D-Flt3-Wt cells was significantly enhanced in FL-treated Pim2-overexpressing cells. This increase was associated with enhanced S-phase cell cycle progression. Pim2-overexpressing cells were resistant to apoptosis induced by growth factor deprivation or treatment with tyrosine kinase inhibitor (PKC412). The Flt3 point mutant D835Y, which is not able to support colony growth of myeloid cells, also induced clonogenic growth in the presence of Pim2. In conclusion, Pim2 is an important target of Flt3-ITD-induced transformation, and overexpression of Pim2 together with Flt3-Wt or D835Y receptor mimics Flt3-ITD-mediated transformation. Pim2 complements with Flt3-Wt signaling to induce proliferation by enhancing G1/S-phase progression of the cell cycle.


Blood | 2011

AML1/ETO induces self-renewal in hematopoietic progenitor cells via the Groucho-related amino-terminal AES protein

Björn Steffen; Markus Knop; Ulla Bergholz; Olesya Vakhrusheva; Miriam Rode; Gabriele Köhler; Marcel-Philipp Henrichs; Etmar Bulk; Sina Hehn; Martin Stehling; Martin Dugas; Nicole Bäumer; Petra Tschanter; Christian Brandts; Steffen Koschmieder; Wolfgang E. Berdel; Hubert Serve; Carol Stocking; Carsten Müller-Tidow

The most frequent translocation t(8;21) in acute myeloid leukemia (AML) generates the chimeric AML1/ETO protein, which blocks differentiation and induces self-renewal in hematopoietic progenitor cells. The underlying mechanisms mediating AML1/ETO-induced self-renewal are largely unknown. Using expression microarray analysis, we identified the Groucho-related amino-terminal enhancer of split (AES) as a consistently up-regulated AML1/ETO target. Elevated levels of AES mRNA and protein were confirmed in AML1/ETO-expressing leukemia cells, as well as in other AML specimens. High expression of AES mRNA or protein was associated with improved survival of AML patients, even in the absence of t(8;21). On a functional level, knockdown of AES by RNAi in AML1/ETO-expressing cell lines inhibited colony formation. Similarly, self-renewal induced by AML1/ETO in primary murine progenitors was inhibited when AES was decreased or absent. High levels of AES expression enhanced formation of immature colonies, serial replating capacity of primary cells, and colony formation in colony-forming unit-spleen assays. These findings establish AES as a novel AML1/ETO-induced target gene that plays an important role in the self-renewal phenotype of t(8;21)-positive AML.


International Journal of Cancer | 2005

Expression patterns of mitotic and meiotic cell cycle regulators in testicular cancer and development

Sven Diederichs; Nicole Bäumer; Nikolaus Schultz; F. Kent Hamra; Mark Schrader; Marie Luise Sandstede; Wolfgang E. Berdel; Hubert Serve; Carsten Müller-Tidow

Mitotic and meiotic cell cycle regulation is essential for normal development and tumor prevention. The underlying molecular mechanisms are not completely characterized. The aim of our analysis was to derive a global expression map of cell cycle regulators in mitosis and meiosis. First, the expression of cyclins, CDKs and CDK inhibitors was determined during postnatal testis maturation in mice using microarrays and quantitative RT‐PCR. The abundance of cyclins A1, B2, K, M4, CDK2, all CDKLs, CDKN2c, CDKN2d and INCA1 increased during testis maturation. In contrast, cyclins A2, B1, D2, G1, G2, CDK1, CDK4 and CDK2AP1 showed a maturation‐associated decrease. Gene expression profiles of isolated germ cells and testicular somatic cells confirmed these results. Second, we determined cyclin expression patterns in human normal and malignant testis samples (n = 36) modeling the reciprocal difference between meiosis and mitosis. Testicular tumors strictly expressed cell cycle regulators identified in mitotically dividing germ cells. Expression of several transcripts was histology‐specific in testicular tumors, providing novel molecular markers and potential therapeutic targets. Taken together, our data provide a comprehensive expression map of cell cycle regulators at the switch between mitosis and meiosis in testis development and in cancerogenesis.


PLOS ONE | 2011

The Inhibitor of Growth Protein 5 (ING5) Depends on INCA1 as a Co-Factor for Its Antiproliferative Effects

Feng Zhang; Nicole Bäumer; Miriam Rode; Ping Ji; Tao Zhang; Wolfgang E. Berdel; Carsten Müller-Tidow

The proteins of the Inhibitor of Growth (ING) family are involved in multiple cellular functions such as cell cycle regulation, apoptosis, and chromatin remodeling. For ING5, its actual role in growth suppression and the necessary partners are not known. In a yeast-two-hybrid approach with human bone marrow derived cDNA, we identified ING5 as well as several other proteins as interaction partners of Inhibitor of cyclin A1 (INCA1) that we previously characterized as a novel interaction partner of cyclin A1/CDK2. ING5 expression in leukemic AML blasts was severely reduced compared to normal bone marrow. In line, ING5 inhibited bone marrow colony formation upon retroviral transduction. However, Inca1 −/− bone marrow colony formation was not suppressed by ING5. In murine embryonic fibroblast (MEF) cells from Inca1+/+ and Inca1−/− mice, overexpression of ING5 suppressed cell proliferation only in the presence of INCA1, while ING5 had no effect in Inca1−/− MEFs. ING5 overexpression induced a delay in S-phase progression, which required INCA1. Finally, ING5 overexpression enhanced Fas-induced apoptosis in Inca1+/+ MEFs, while Inca1−/− MEFs were protected from Fas antibody-induced apoptosis. Taken together, these results indicate that ING5 is a growth suppressor with suppressed expression in AML whose functions depend on its interaction with INCA1.


Journal of Biological Chemistry | 2011

Inhibitor of Cyclin-dependent Kinase (CDK) Interacting with Cyclin A1 (INCA1) Regulates Proliferation and Is Repressed by Oncogenic Signaling

Nicole Bäumer; Lara Tickenbrock; Petra Tschanter; Lisa Lohmeyer; Sven Diederichs; Sebastian Bäumer; Boris V. Skryabin; Feng Zhang; Shuchi Agrawal-Singh; Gabriele Köhler; Wolfgang E. Berdel; Hubert Serve; Steffen Koschmieder; Carsten Müller-Tidow

The cell cycle is driven by the kinase activity of cyclin·cyclin-dependent kinase (CDK) complexes, which is negatively regulated by CDK inhibitor proteins. Recently, we identified INCA1 as an interaction partner and a substrate of cyclin A1 in complex with CDK2. On a functional level, we identified a novel cyclin-binding site in the INCA1 protein. INCA1 inhibited CDK2 activity and cell proliferation. The inhibitory effects depended on the cyclin-interacting domain. Mitogenic and oncogenic signals suppressed INCA1 expression, whereas it was induced by cell cycle arrest. We established a deletional mouse model that showed increased CDK2 activity in spleen with altered spleen architecture in Inca1−/− mice. Inca1−/− embryonic fibroblasts showed an increase in the fraction of S-phase cells. Furthermore, blasts from acute lymphoid leukemia and acute myeloid leukemia patients expressed significantly reduced INCA1 levels highlighting its relevance for growth control in vivo. Taken together, this study identifies a novel CDK inhibitor with reduced expression in acute myeloid and lymphoid leukemia. The molecular events that control the cell cycle occur in a sequential process to ensure a tight regulation, which is important for the survival of a cell and includes the detection and repair of genetic damage and the prevention of uncontrolled cell division.

Collaboration


Dive into the Nicole Bäumer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sven Diederichs

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Ping Ji

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge