Network


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

Hotspot


Dive into the research topics where Simon J. Baumgart is active.

Publication


Featured researches published by Simon J. Baumgart.


EMBO Reports | 2016

Loss of CHD1 causes DNA repair defects and enhances prostate cancer therapeutic responsiveness

Vijayalakshmi Kari; Wael Y. Mansour; Sanjay Kumar Raul; Simon J. Baumgart; Andreas Mund; Marian Grade; Hüseyin Sirma; Ronald Simon; Hans Will; Matthias Dobbelstein; Ekkehard Dikomey; Steven A. Johnsen

The CHD1 gene, encoding the chromo‐domain helicase DNA‐binding protein‐1, is one of the most frequently deleted genes in prostate cancer. Here, we examined the role of CHD1 in DNA double‐strand break (DSB) repair in prostate cancer cells. We show that CHD1 is required for the recruitment of CtIP to chromatin and subsequent end resection during DNA DSB repair. Our data support a role for CHD1 in opening the chromatin around the DSB to facilitate the recruitment of homologous recombination (HR) proteins. Consequently, depletion of CHD1 specifically affects HR‐mediated DNA repair but not non‐homologous end joining. Together, we provide evidence for a previously unknown role of CHD1 in DNA DSB repair via HR and show that CHD1 depletion sensitizes cells to PARP inhibitors, which has potential therapeutic relevance. Our findings suggest that CHD1 deletion, like BRCA1/2 mutation in ovarian cancer, may serve as a marker for prostate cancer patient stratification and the utilization of targeted therapies such as PARP inhibitors, which specifically target tumors with HR defects.


Nucleic Acids Research | 2017

BRD4 localization to lineage-specific enhancers is associated with a distinct transcription factor repertoire

Zeynab Najafova; Roberto Tirado-Magallanes; Malayannan Subramaniam; Tareq Hossan; Geske Schmidt; Sankari Nagarajan; Simon J. Baumgart; Vivek Kumar Mishra; Upasana Bedi; Eric Hesse; Stefan Knapp; John R. Hawse; Steven A. Johnsen

Proper temporal epigenetic regulation of gene expression is essential for cell fate determination and tissue development. The Bromodomain-containing Protein-4 (BRD4) was previously shown to control the transcription of defined subsets of genes in various cell systems. In this study we examined the role of BRD4 in promoting lineage-specific gene expression and show that BRD4 is essential for osteoblast differentiation. Genome-wide analyses demonstrate that BRD4 is recruited to the transcriptional start site of differentiation-induced genes. Unexpectedly, while promoter-proximal BRD4 occupancy correlated with gene expression, genes which displayed moderate expression and promoter-proximal BRD4 occupancy were most highly regulated and sensitive to BRD4 inhibition. Therefore, we examined distal BRD4 occupancy and uncovered a specific co-localization of BRD4 with the transcription factors C/EBPb, TEAD1, FOSL2 and JUND at putative osteoblast-specific enhancers. These findings reveal the intricacies of lineage specification and provide new insight into the context-dependent functions of BRD4.


Genome Biology | 2017

RNF40 regulates gene expression in an epigenetic context-dependent manner

Wanhua Xie; Sankari Nagarajan; Simon J. Baumgart; Robyn Laura Kosinsky; Zeynab Najafova; Vijayalakshmi Kari; Magali Hennion; Daniela Indenbirken; Stefan Bonn; Adam Grundhoff; Florian Wegwitz; Ahmed Mansouri; Steven A. Johnsen

BackgroundMonoubiquitination of H2B (H2Bub1) is a largely enigmatic histone modification that has been linked to transcriptional elongation. Because of this association, it has been commonly assumed that H2Bub1 is an exclusively positively acting histone modification and that increased H2Bub1 occupancy correlates with increased gene expression. In contrast, depletion of the H2B ubiquitin ligases RNF20 or RNF40 alters the expression of only a subset of genes.ResultsUsing conditional Rnf40 knockout mouse embryo fibroblasts, we show that genes occupied by low to moderate amounts of H2Bub1 are selectively regulated in response to Rnf40 deletion, whereas genes marked by high levels of H2Bub1 are mostly unaffected by Rnf40 loss. Furthermore, we find that decreased expression of RNF40-dependent genes is highly associated with widespread narrowing of H3K4me3 peaks. H2Bub1 promotes the broadening of H3K4me3 to increase transcriptional elongation, which together lead to increased tissue-specific gene transcription. Notably, genes upregulated following Rnf40 deletion, including Foxl2, are enriched for H3K27me3, which is decreased following Rnf40 deletion due to decreased expression of the Ezh2 gene. As a consequence, increased expression of some RNF40-“suppressed” genes is associated with enhancer activation via FOXL2.ConclusionTogether these findings reveal the complexity and context-dependency whereby one histone modification can have divergent effects on gene transcription. Furthermore, we show that these effects are dependent upon the activity of other epigenetic regulatory proteins and histone modifications.


Cancer Research | 2017

Krüppel-like Transcription Factor KLF10 Suppresses TGFβ-Induced Epithelial-to-Mesenchymal Transition via a Negative Feedback Mechanism

Vivek Kumar Mishra; Malayannan Subramaniam; Vijayalakshmi Kari; Kevin S. Pitel; Simon J. Baumgart; Ryan M. Naylor; Sankari Nagarajan; Florian Wegwitz; V Ellenrieder; John R. Hawse; Steven A. Johnsen

TGFβ-SMAD signaling exerts a contextual effect that suppresses malignant growth early in epithelial tumorigenesis but promotes metastasis at later stages. Longstanding challenges in resolving this functional dichotomy may uncover new strategies to treat advanced carcinomas. The Krüppel-like transcription factor, KLF10, is a pivotal effector of TGFβ/SMAD signaling that mediates antiproliferative effects of TGFβ. In this study, we show how KLF10 opposes the prometastatic effects of TGFβ by limiting its ability to induce epithelial-to-mesenchymal transition (EMT). KLF10 depletion accentuated induction of EMT as assessed by multiple metrics. KLF10 occupied GC-rich sequences in the promoter region of the EMT-promoting transcription factor SLUG/SNAI2, repressing its transcription by recruiting HDAC1 and licensing the removal of activating histone acetylation marks. In clinical specimens of lung adenocarcinoma, low KLF10 expression associated with decreased patient survival, consistent with a pivotal role for KLF10 in distinguishing the antiproliferative versus prometastatic functions of TGFβ. Our results establish that KLF10 functions to suppress TGFβ-induced EMT, establishing a molecular basis for the dichotomy of TGFβ function during tumor progression. Cancer Res; 77(9); 2387-400. ©2017 AACR.


Stem Cells | 2016

Histone Chaperone SSRP1 is Essential for Wnt Signaling Pathway Activity During Osteoblast Differentiation

Tareq Hossan; Sankari Nagarajan; Simon J. Baumgart; Wanhua Xie; Roberto Tirado Magallanes; Céline Hernandez; Pierre-Marie Chiaroni; Daniela Indenbirken; Melanie Spitzner; Morgane Thomas-Chollier; Marian Grade; Denis Thieffry; Adam Grundhoff; Florian Wegwitz; Steven A. Johnsen

Cellular differentiation is accompanied by dramatic changes in chromatin structure which direct the activation of lineage‐specific transcriptional programs. Structure‐specific recognition protein‐1 (SSRP1) is a histone chaperone which is important for chromatin‐associated processes such as transcription, DNA replication and repair. Since the function of SSRP1 during cell differentiation remains unclear, we investigated its potential role in controlling lineage determination. Depletion of SSRP1 in human mesenchymal stem cells elicited lineage‐specific effects by increasing expression of adipocyte‐specific genes and decreasing the expression of osteoblast‐specific genes. Consistent with a role in controlling lineage specification, transcriptome‐wide RNA‐sequencing following SSRP1 depletion and the induction of osteoblast differentiation revealed a specific decrease in the expression of genes involved in biological processes related to osteoblast differentiation. Importantly, we observed a specific downregulation of target genes of the canonical Wnt signaling pathway, which was accompanied by decreased nuclear localization of active β‐catenin. Together our data uncover a previously unknown role for SSRP1 in promoting the activation of the Wnt signaling pathway activity during cellular differentiation. Stem Cells 2016;34:1369–1376


The Journal of Urology | 2017

MP28-03 ALTERATION OF METASTATIC BEHAVIOR BY SHRNA MEDIATED KNOCKDOWN (KD) OF CHD1 IN HUMAN PROSTATE XENOGRAFT TUMORS AND CLINICAL OUTCOME OF PATIENTS WITH CHD1 DELETION

Su Jung Oh; Derya Tilki; Christiane Matuszcak; Pierre Tennstedt; Simon J. Baumgart; Steven A. Johnsen; Hüseyin Sirma; Ronald Simon; Tobias Lange

INTRODUCTION AND OBJECTIVES: Epidemiologic studies have reported an association between frequent consumption of welldone cooked meats and prostate cancer (PC) risk. Charred red meat and cooked processed meats are known to contain heterocyclic aromatic amine (HAA) carcinogens, such as 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) the most mass abundant HAA, and are linked to PC development in a rodent model. However, unambiguous physiochemical markers of DNA damage from these meat-derived carcinogens have not been identified in human samples to support the paradigm of HAA induced human prostate carcinogenesis. METHODS: Thirty-five men with biopsy proven intermediate to high-risk PC underwent radical prostatectomy at University of Minnesota from Dec 2015-Aug 2016. After prostatectomy, both tumor bearing tissue and non-tumor bearing adjacent fresh tissue was analyzed for DNA adducts using a highly sensitive nano-LC-Orbitrap mass spectrometry method. We also analyzed formalin fixed paraffin embedded (FFPE) tissues from each patient. RESULTS: Median age of the men with PC was 65 (range 4578). Pathology demonstrated the following Gleason Scores (GS) and pathologic staging: GS1⁄4 6 in 1 patient (2.8%), GS1⁄47 in 28 patients (80%) and GS1⁄48-10 in 6 patients (17%) and 16 men (46%) were stage 2 and 19 men were stage 3 (54%). The PhIP DNA adduct was identified in 11 out of 35 patients, at levels ranging from 2 to 120 adducts per 109 nucleotides. PhIP DNA adducts also were recovered quantitatively from FFPE tissues. CONCLUSIONS: Our data provide support to the epidemiological observations implicating PhIP as a DNA damaging agent that may contribute to the etiology of PC in humans. FFPE tissues can be used as a tissue source in DNA-adduct biomarker research using our mass spectrometry method.


Nucleic Acids Research | 2017

CHD1 regulates cell fate determination by activation of differentiation-induced genes

Simon J. Baumgart; Zeynab Najafova; Tareq Hossan; Wanhua Xie; Sankari Nagarajan; Vijayalakshmi Kari; Nicholas Ditzel; Moustapha Kassem; Steven A. Johnsen

Abstract The coordinated temporal and spatial activation of gene expression is essential for proper stem cell differentiation. The Chromodomain Helicase DNA-binding protein 1 (CHD1) is a chromatin remodeler closely associated with transcription and nucleosome turnover downstream of the transcriptional start site (TSS). In this study, we show that CHD1 is required for the induction of osteoblast-specific gene expression, extracellular-matrix mineralization and ectopic bone formation in vivo. Genome-wide occupancy analyses revealed increased CHD1 occupancy around the TSS of differentiation-activated genes. Furthermore, we observed that CHD1-dependent genes are mainly induced during osteoblast differentiation and are characterized by higher levels of CHD1 occupancy around the TSS. Interestingly, CHD1 depletion resulted in increased pausing of RNA Polymerase II (RNAPII) and decreased H2A.Z occupancy close to the TSS, but not at enhancer regions. These findings reveal a novel role for CHD1 during osteoblast differentiation and provide further insights into the intricacies of epigenetic regulatory mechanisms controlling cell fate determination.


Cancer Research | 2016

Abstract A03: Krüppel-like Transcription Factor-10 (KLF10) suppresses the TGFβ-induced epithelial-to-mesenchymal transition

Vivek Kumar Mishra; Vijayalakshmi Kari; Malayannan Subramaniam; Simon J. Baumgart; Sankari Nagarajan; Florian Wegwitz; Thomas C. Spelsberg; John R. Hawse; Steven A. Johnsen

The Transforming Growth Factor-β (TGFβ)/SMAD signaling pathway can function as either a tumor suppressor or metastasis promoter during tumor progression. In normal epithelial cells and early stages of epithelial tumorigenesis TGFβ functions as a tumor suppressor to decrease cell proliferation or induce apoptosis. However, during malignant progression tumor cells no longer respond to the anti-proliferative effects of TGFβ, but instead undergo an epithelial-to-mesenchymal transition (EMT) whereby cells acquire a migratory and invasive phenotype which promotes tumor metastasis. Resolution of the dichotomy in TGFβ function and a further understanding of its tumor suppressor and metastasis promoting functions may uncover new strategies for the treatment of epithelial cancers. In previous studies we demonstrated an important role of the TGFβ-Inducible Early Gene-1 (TIEG1)/Kruppel-like Factor-10 (KLF10) as a central regulator of TGFβ/SMAD signaling and the anti-proliferative functions of TGFβ. In this study we examined the role of KLF10 in controlling the TGFβ-induced EMT in human lung adenocarcinoma cells. We show that depletion of KLF10 results in a more pronounced induction of EMT in human A549 lung adenocarcinoma cells as assessed by quantitative real-time PCR, changes in cellular morphology and immunofluorescence microscopy, and transcriptome-wide (RNA-seq) analyses. Moreover, chromatin immunoprecipitation (ChIP) and chromatin immunoprecipitation-sequencing (ChIP-seq) analysis shows that KLF10 directly binds to GC-rich sequences in the promoter region of the EMT-promoting transcription factor SLUG/SNAI2 to repress its transcription. Consistent with these findings, an analysis of KLF10 and SNAI2 expression in lung cancer revealed that while KLF10 levels are decreased in lung cancer vs. normal samples, SNAI2 levels are increased. Additional ChIP studies showed that KLF10 recruits HDAC1 to the SNAI2 promoter and is required for the removal of activating histone acetylation marks. These findings reveal a previously unknown function of KLF10 in suppressing TGFβ-induced EMT and represent a significant advancement in the understanding the molecular dichotomy of TGFβ function during tumor progression. Citation Format: Vivek Kumar Mishra, Vijayalakshmi Kari, Malayannan Subramaniam, Simon J. Baumgart, Sankari Nagarajan, Florian Wegwitz, Thomas C. Spelsberg, John R. Hawse, Steven A. Johnsen. Kruppel-like Transcription Factor-10 (KLF10) suppresses the TGFβ-induced epithelial-to-mesenchymal transition. [abstract]. In: Proceedings of the AACR Special Conference on Chromatin and Epigenetics in Cancer; Sep 24-27, 2015; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2016;76(2 Suppl):Abstract nr A03.


Cancer Research | 2018

Abstract 1917: CHD1 depletion leads to androgen-independent growth of prostate cancer cells lines and regulates the TGFβ system in an EZH2-dependent manner

Simon J. Böcker; Simon J. Baumgart; Annabell Roth; Vijayalakshmi Kari; Steven A. Johnsen; Oliver Hahn


Cancer Research | 2018

Abstract 1795: Darolutamide demonstrates greater in vivo activity than enzalutamide in prostate cancer model with therapy-resistant androgen receptor mutation

Bernard Haendler; Tatsuo Sugawara; Simon J. Baumgart; Pascale Lejeune

Collaboration


Dive into the Simon J. Baumgart's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tareq Hossan

University of Göttingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wanhua Xie

University of Göttingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge