Network


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

Hotspot


Dive into the research topics where Maren Kaufmann is active.

Publication


Featured researches published by Maren Kaufmann.


International Journal of Cancer | 1999

Widespread intraspecies cross‐contamination of human tumor cell lines arising at source

Roderick A. F. MacLeod; Wilhelm G. Dirks; Yoshinobu Matsuo; Maren Kaufmann; Herbert Milch; Hans G. Drexler

We present a panoptic survey of cell line cross‐contamination (CLCC) among original stocks of human cell lines, investigated using molecular genetic methods. The survey comprised 252 consecutive human cell lines, almost exclusively tumor‐derived, submitted by their originators to the DSMZ and 5 additional cell repositories (CRs), using a combination of DNA profiling (4‐locus minisatellite and multilocus microsatellite probes) and molecular cytogenetics, exploiting an interactive database (http://www.dsmz.de/). Widespread high levels of cross‐contaminants (CCs) were uncovered, affecting 45 cell lines (18%) supplied by 27 of 93 originators (29%). Unlike previous reports, most CCs (42/45) occurred intraspecies, a discrepancy attributable to improved detection of the more insidious intraspecies CCs afforded by molecular methods. The most prolific CCs were classic tumor cell lines, the numbers of CCs they caused being as follows: HeLa (n = 11), T‐24 (n = 4), SK‐HEP‐1 (n = 4), U‐937 (n = 4) and HT‐29 (n = 3). All 5 supposed instances of spontaneous immortalization of normal cells were spurious, due to CLCC, including ECV304, the most cited human endothelial cell line. Although high, our figure for CCs at the source sets a lower limit only as (i) many older tumor cell lines were unavailable for comparison and (ii) circulating cell lines are often obtained indirectly, rather than via originators or CRs. The misidentified cell lines reported here have already been unwittingly used in several hundreds of potentially misleading reports, including use as inappropriate tumor models and subclones masquerading as independent replicates. We believe these findings indicate a grave and chronic problem demanding radical measures, to include extra controls over cell line authentication, provenance and availability. Int. J. Cancer 83:555–563, 1999.


Nature Protocols | 2007

Cytogenetic harvesting of commonly used tumor cell lines

Roderick A. F. MacLeod; Maren Kaufmann; Hans G. Drexler

Tumor cell lines are widely used both as disease models and, increasingly, as genomic resources for the ascertainment of new cancer genes. Cytogenetic analysis remains a major route to uncovering the cancer genome. However, cancer cell lines vary inexplicably in their harvesting preferences, which must, therefore, be determined by trial and error. This article describes harvesting protocols optimized empirically for 550 commonly used, mainly human, cancer cell lines together with evidence-based procedures to assist in determining conditions for unlisted cell lines and subsidiary protocols for cytogenetic analysis using G-banding and fluorescence in situ hybridization.


Genes, Chromosomes and Cancer | 2003

Activation of HOX11L2 by juxtaposition with 3′-BCL11B in an acute lymphoblastic leukemia cell line (HPB-ALL) with t(5;14)(q35;q32.2)

Roderick A. F. MacLeod; Stefan Nagel; Maren Kaufmann; Johannes W.G. Janssen; Hans G. Drexler

Cytogenetic analysis of a pediatric T‐cell acute lymphoblastic leukemia (ALL) cell line (HPB‐ALL) revealed the cryptic t(5;14)(q35;q32.2), recently found in 15–20% pediatric T‐ALL patients, with 5q35 and 14q32.2 breakpoints at 5′‐HOX11L2 and 3′‐BCL11B, respectively. Expression of both BCL11B, which is hematologically restricted to T cells, and HOX11L2 was detected, whereas adjacent genes at 5q35 (RANBP17) and 14q32 (VRK1, HSU88895) were not dysregulated. Apparently, t(5;14)(q35;q32.2) serves to activate HOX11L2 by juxtaposition with elements downstream of BCL11B, implying a novel dysregulatory mechanism comparable to TCRA/D or IGH juxtaposition. As well as providing molecular cytogenetic documentation of t(5;14) ALL, this report validates HPB‐ALL cells for investigating this important new disease entity in vitro.


Leukemia | 2008

Amplification at 7q22 targets cyclin-dependent kinase 6 in T-cell lymphoma

Stefan Nagel; Ellen Leich; Hilmar Quentmeier; Corinna Meyer; Maren Kaufmann; Hans G. Drexler; A Zettl; Andreas Rosenwald; Roderick A. F. MacLeod

Recurrent chromosomal aberrations in hematopoietic tumors target genes involved in pathogenesis. Their identification and functional characterization are therefore important for the establishment of rational therapies. Here, we investigated genomic amplification at 7q22 in the T-cell lymphoma cell line SU-DHL-1 belonging to the subtype of anaplastic large-cell lymphoma (ALCL). Cytogenetic analysis mapped this amplicon to 86–95 Mb. Copy-number determination quantified the amplification level at 5- to 6-fold. Expression analysis of genes located within this region identified cyclin-dependent kinase 6 (CDK6) as a potential amplification target. In comparison with control cell lines, SU-DHL-1 expressed considerably higher levels of CDK6. Functionally, SU-DHL-1 cells exhibited reduced sensitivity to rapamycin treatment, as indicated by cell growth and cell cycle analysis. Rapamycin reportedly inhibits degradation of the CDK inhibitor p27 with concomitant downregulation of cyclin D3, implying a proliferative advantage for CDK6 overexpression. Amplification of the CDK6 locus was analyzed in primary T-cell lymphoma samples and, while detected infrequently in those classified as ALCL (1%), was detected in 23% of peripheral T-cell lymphomas not otherwise specified. Taken together, analysis of the 7q22 amplicon identified CDK6 as an important cell cycle regulator in T-cell lymphomas, representing a novel potential target for rational therapy.


Leukemia | 2005

HLXB9 activates IL6 in Hodgkin lymphoma cell lines and is regulated by PI3K signalling involving E2F3.

Stefan Nagel; Michaela Scherr; Hilmar Quentmeier; Maren Kaufmann; Margarete Zaborski; Hans G. Drexler; Roderick A. F. MacLeod

Multiple cytokines are secreted by Hodgkin lymphoma (HL) cells, notably interleukin-6 (IL6), which is believed to play a significant pathobiological role in this and certain other tumors. Previous work on prostate carcinoma cells has shown that IL6 expression is activated therein by the homeodomain protein GBX2, which we found to be absent in HL cells. Instead, we observed expression of a closely related gene, HLXB9, albeit restricted to HL cells coexpressing IL6. Treatment of HL cell lines with antisense-oligonucleotides directed against HLXB9, forced expression of recombinant HLXB9, and analysis of reporter gene constructs containing IL6 promoter sequences all confirmed the potential of HLXB9 to drive expression of IL6. Chromosomal rearrangements of the HLXB9 locus at 7q36 were not detected in HL cells unlike AML subsets expressing HLXB9. However, inhibition of certain signal transduction pathways revealed that the phosphatidylinositol 3 kinase (PI3K) pathway contributes to HLXB9 expression. AKT/phospho-AKT analysis revealed constitutively active PI3K signalling in HL cell lines. Downstream analysis of PI3K revealed that E2F3 may mediate activation of HLXB9. Taken together, our data show that the PI3K signalling pathway in HL cells is constitutively activated and promotes HLXB9 expression, probably via E2F3, thereby enhancing malignant expression of IL6.


Leukemia | 2008

MEF2C is activated by multiple mechanisms in a subset of T-acute lymphoblastic leukemia cell lines

Stefan Nagel; Corinna Meyer; Hilmar Quentmeier; Maren Kaufmann; Hans G. Drexler; Roderick A. F. MacLeod

In T-cell acute lymphoblastic leukemia (T-ALL) the cardiac homeobox gene NKX2-5 (at 5q35) is variously deregulated by regulatory elements coordinating with BCL11B (at 14q32.2), or the T-cell receptor gene TRD (at 14q11.2), respectively. NKX2-5 is normally expressed in developing spleen and heart, regulating fundamental processes, including differentiation and survival. In this study we investigated whether NKX2-5 expression in T-ALL cell lines reactivates these embryonal pathways contributing to leukemogenesis. Among 18 known targets analyzed, we identified three genes regulated by NKX2-5 in T-ALL cells, including myocyte enhancer factor 2C (MEF2C). Knockdown and overexpression assays confirmed MEF2C activation by NKX2-5 at both the RNA and protein levels. Direct interactions between NKX2-5 and GATA3 as indicated by co-immunoprecipitation data may contribute to MEF2C regulation. In T-ALL cell lines LOUCY and RPMI-8402 MEF2C expression was correlated with a 5q14 deletion, encompassing noncoding proximal gene regions. Fusion constructs with green fluorescent protein permitted subcellular detection of MEF2C protein in nuclear speckles interpretable as repression complexes. MEF2C consistently inhibits expression of NR4A1/NUR77, which regulates apoptosis via BCL2 transformation. Taken together, our data identify distinct mechanisms underlying ectopic MEF2C expression in T-ALL, either as a downstream target of NKX2-5, or via chromosomal aberrations deleting proximal gene regions.


Leukemia Research | 2002

Multicolor-FISH analysis of a natural killer cell line (NK-92).

Roderick A. F. MacLeod; Stefan Nagel; Maren Kaufmann; Karin M. Greulich-Bode; Hans G. Drexler

Relatively little is known about the cytogenetics of natural killer (NK) neoplasms, and the emergence of recurrent structural alterations involving specific chromosomal breakpoints is still in its infancy. This gap has doubtless hampered identification of the oncogene alterations posited to underly NK tumors. We describe in detail the cytogenetic rearrangements present in a cytotoxic NK cell line (NK-92) established from a patient with large granular lymphocyte (LGL) lymphoma. The NK-92 cell line is one of very few cytotoxic NK cell lines described and the first to be used clinically. Cytogenetic analysis was performed independently using two multicolor-fluorescence in situ hybridization (M-FISH) systems, the first M-FISH study of a cell line derived from a NK neoplasm. Several non-random cytogenetic features previously reported in NK cells were, thus, identified, including rearrangements of chromosomes 7 and 17, along with breakpoints at 11q23, 12q12 and 8p22/23. FISH revealed that NK-92 cells carry multiple rearrangements with distinct breakpoints at 8p resembling those previously described in NK lymphoma. Our data strengthen the claim of NK-92 to model NK neoplasms and highlight this cell line as a potential resource for mining relevant oncogenic changes therein.


Molecular Cancer | 2010

Polycomb repressor complex 2 regulates HOXA9 and HOXA10, activating ID2 in NK/T-cell lines

Stefan Nagel; Letizia Venturini; Victor E. Marquez; Corinna Meyer; Maren Kaufmann; Michaela Scherr; Roderick A. F. MacLeod; Hans G. Drexler

BackgroundNK- and T-cells are closely related lymphocytes, originating from the same early progenitor cells during hematopoiesis. In these differentiation processes deregulation of developmental genes may contribute to leukemogenesis. Here, we compared expression profiles of NK- and T-cell lines for identification of aberrantly expressed genes in T-cell acute lymphoblastic leukemia (T-ALL) which physiologically regulate the differentiation program of the NK-cell lineage.ResultsThis analysis showed high expression levels of HOXA9, HOXA10 and ID2 in NK-cell lines in addition to T-cell line LOUCY, suggesting leukemic deregulation therein. Overexpression experiments, chromatin immuno-precipitation and promoter analysis demonstrated that HOXA9 and HOXA10 directly activated expression of ID2. Concomitantly elevated expression levels of HOXA9 and HOXA10 together with ID2 in cell lines containing MLL translocations confirmed this form of regulation in both ALL and acute myeloid leukemia. Overexpression of HOXA9, HOXA10 or ID2 resulted in repressed expression of apoptosis factor BIM. Furthermore, profiling data of genes coding for chromatin regulators of homeobox genes, including components of polycomb repressor complex 2 (PRC2), indicated lacking expression of EZH2 in LOUCY and exclusive expression of HOP in NK-cell lines. Subsequent treatment of T-cell lines JURKAT and LOUCY with DZNep, an inhibitor of EZH2/PRC2, resulted in elevated and unchanged HOXA9/10 expression levels, respectively. Moreover, siRNA-mediated knockdown of EZH2 in JURKAT enhanced HOXA10 expression, confirming HOXA10-repression by EZH2. Additionally, profiling data and overexpression analysis indicated that reduced expression of E2F cofactor TFDP1 contributed to the lack of EZH2 in LOUCY. Forced expression of HOP in JURKAT cells resulted in reduced HOXA10 and ID2 expression levels, suggesting enhancement of PRC2 repression.ConclusionsOur results show that major differentiation factors of the NK-cell lineage, including HOXA9, HOXA10 and ID2, were (de)regulated via PRC2 which therefore contributes to T-cell leukemogenesis.


Leukemia Research | 2010

Multiple mechanisms induce ectopic expression of LYL1 in subsets of T-ALL cell lines

Stefan Nagel; Letizia Venturini; Corinna Meyer; Maren Kaufmann; Michaela Scherr; Hans G. Drexler; Roderick A. F. MacLeod

Basic helix-loop-helix (bHLH) transcription factors are essential for lymphocytic differentiation. Here, we have analyzed the complete bHLH family in T-cell acute lymphoblastic leukemia cell lines by expression profiling. Differential expression was detected for BHLHB2, HES1, HES4, HEY1, ID1, ID2, ID3, LYL1 and TAL1, highlighting dysregulation of family members with inhibitory activity. Subsequently we focused on the mechanisms responsible for aberrant expression of LYL1 in comparison to TAL1. Quantitative genomic PCR indicated microdeletions upstream of both, TAL1 and LYL1, targeting STIL/SIL and TRMT1, respectively. Additionally, one LYL1-expressing cell line exhibited amplification of TRMT1. While deletion of STIL correlated with expression of the STIL-TAL1 fusion transcript, no TRMT-LYL1 fusion transcripts were detected in parallel with genomic rearrangements thereof. Sequence analysis of the LYL1 promoter region revealed potential binding sites for transcription factors HOXA10, LMO2 and NKX2-5. Overexpression analysis, reporter gene assays and chromatin immuno-precipitation confirmed their activating impact on LYL1 expression. In conclusion, we identified multiple mechanisms which activate LYL1 in leukemic cells, including structural genomic alterations, namely microdeletion or amplification, together with the involvement of prominent oncogenic transcription factors.


Leukemia | 2008

T(3;7)(q27;q32) fuses BCL6 to a non-coding region at FRA7H near miR-29.

Björn Schneider; Stefan Nagel; Maren Kaufmann; S Winkelmann; J Bode; Hans G. Drexler; Roderick A. F. MacLeod

Zucker A, Luberto C et al. The CLN9 protein, a regulator of dihydroceramide synthase. J Biol Chem 2006; 281: 2784–2794. 7 Zheng W, Kollmeyer J, Symolon H, Momin A, Munter E, Wang E et al. Ceramides and other bioactive sphingolipid backbones in health and disease: lipidomic analysis, metabolism and roles in membrane structure, dynamics, signaling and autophagy. Biochim Biophys Acta 2006; 1758: 1864–1884. 8 Kraveka JM, Li L, Szulc ZM, Bielawski J, Ogretmen B, Hannun YA et al. Involvement of the dihydroceramide desaturase in cell cycle progression in human neuroblastoma cells. J Biol Chem 2007; 282: 16718–16728.

Collaboration


Dive into the Maren Kaufmann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hans G. Drexler

Deutsche Sammlung von Mikroorganismen und Zellkulturen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Björn Schneider

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge