Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 2-2
    Abstract: Differentiation of hematopoietic stem cells follows a hierarchical program of transcriptional-regulated events. We here identify SMARCD2 (Swi/Snf-related matrix associated actin dependent regulator of chromatin, subfamily D, member 2) as critical regulator of myelopoiesis in humans, mice, and zebrafish. We studied four patients from three unrelated pedigrees presenting with a novel syndromatic phenotype comprising congenital neutropenia, specific granule deficiency, susceptibility to myelodysplasia with excess of blasts, and various skeletal anomalies. All patients had homozygous loss-of-function mutations in SMARCD2. In contrast to wildtype alleles, the variant alleles did not give rise to proteins with capacity to interact with the SWI/SNF subunits BRG1, BAF170, BAF155, and BAF47, as shown by co-immunoprecipitation experiments. In vitro, knockdown of SMARCD2 in promyelocytic NB4 cells, differentiated in the presence of ATRA, led to decreased expression of genes encoding the primary granule proteins cathelicidin (CAMP) and alpha-1-antitrypsin (AAT) as well as specific granule proteins matrix metalloproteinase-8 (MMP8), transcobalamin (TCN1) and lactoferrin (LTF). This phenotype is reminiscent of patients with specific granule deficiency, characterized by mutations in CEBPE, a known transcription factor controlling terminal neutrophil development. We therefore hypothesized that SMARCD2 may act via CEBPe and performed immunoprecipitation studies in transfected cells. Upon pull-down of SMARCD2, CEPBE could be detected, and vice versa, suggesting that both proteins physically interact to control transcriptional networks. To interrogate effects of SMARCD2 deficiency on global chromatin accessibility we made use of ATAC sequencing of undifferentiated and ATRA-differentiated NB4 cells and compared this data with comprehensive RNA-sequencing results. A specific subset of genes was found deregulated in both assays, affecting vesicular trafficking, migration and signalling pathways. To validate a role for SMARCD2 in hematopoiesis in vivo, we generated murine and zebrafish model systems. We generated Smarcd2-/- mice by injection of Smarcd2+/- murine ES cells into blastocysts, transfer into pseudo-pregnant mice and interbreeding of heterozygous Smarcd2+/- offsprings. The mutant allele was inherited in a Mendelian fashion but no viable mice were born. 14.5dpc embryos were characterized by anemia and reduced size compared to their littermates. Analysis of fetal liver hematopoiesis revealed a complete absence of CD11b+Gr1+ and CD11b+Ly6c+ cells, whereas the number of LSK stem cells was not affected. Futhermore, Smarcd2-/- embryos showed aberrations in erythroid cells such as extensive anisocytosis, multinucleated cells, and perturbed mitosis. In cytokine-driven colony forming unit assays, GM-CSF, M-CSF, and G-CSF induced myeloid cell differentiation was decreased. Transcriptional profiling of LSK stem cells revealed a striking dysbalance affecting genes involved in signaling pathways and host defence, including CEBPE-dependent genes. Among a total of 12362 detected genes, we found 4290 to be differentially expressed (DESeq2, FDR 〈 10%). Interestingly, the majority (79%) of the 605 genes with a relatively large difference (fold-change 〉 1.4, FDR 〈 1%) were up- and not downregulated. Next, we generated three Smarcd2-deficient zebrafish models using a) morpholino-mediated knockdown in Tg(mpx:EGFP)i114 and Tg(lyz:dsRed)nz50 strains of the orthologous gene or b) Crisp/Cas9-mediated genomic engineering of this locus in Tg(mpx:EGFP)i114. In all models, the numbers of neutrophil granulocytes were significantly reduced. We conclude that SMARCD2 is a critical factor orchestrating transcriptional networks controlling hematopoiesis across species, in particular regulation and maintenance of neutrophil differentation and prevention of leukemogenesis. Disclosures Abboud: Novartis: Honoraria; MAST Therapeutics: Research Funding; Eli Lilly and Company: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Nature Genetics, Springer Science and Business Media LLC, Vol. 49, No. 5 ( 2017-05), p. 742-752
    Type of Medium: Online Resource
    ISSN: 1061-4036 , 1546-1718
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2017
    detail.hit.zdb_id: 1494946-5
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Cell, Elsevier BV, Vol. 30, No. 6 ( 2016-12), p. 849-862
    Type of Medium: Online Resource
    ISSN: 1535-6108
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2016
    detail.hit.zdb_id: 2074034-7
    detail.hit.zdb_id: 2078448-X
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: BMC Public Health, Springer Science and Business Media LLC, Vol. 19, No. 1 ( 2019-12)
    Abstract: Health care employees in Germany and worldwide are exposed to a variety of stressors. However, most of the hospitals in Germany lack a systematic workplace health management. Thus, this study aims at the evaluation of the effects of a behavioural as well as organisational (´complex´) intervention on the mental health and well-being of hospital staff. Methods Mental health in the hospital workplace (SEElische GEsundheit am Arbeitsplatz KrankeNhaus – SEEGEN) is an unblinded, multi-centred cluster-randomised open trial with two groups (intervention group (IG) and waitlist control group (CG)). Study participants in the intervention clusters will receive the complex intervention; study participants in the waitlist control clusters will receive the complex intervention after the last follow-up measurement. The intervention consists of five behavioural and organisational intervention modules that are specifically tailored to hospital employees at different hierarchical and functional levels. Hospital staff may select one specific module according to their position and specific needs or interests. Towards the end of the intervention roundtable discussions with representatives from all professional groups will be held to facilitate organisational change. Primary outcome is the change in emotional and cognitive strain in the working environment, from baseline (T0) to 6 month-follow up (T1), between IG and CG. In addition, employees who do not participate in the modules are included in the trial by answering shorter questionnaires (cluster participants). Furthermore, using mixed methods, a process evaluation will identify uptake of the intervention, and mediators and moderators of the effect. Discussion There seems to be growing psychological strain on people working in the health care sector worldwide. This study will examine whether investing directly in the hospital staff and their interpersonal relationship may lead to measurable benefits in subjective well-being at the workplace and improved economic performance indicators of the hospital. In case of a positive outcome, health promotion strategies looking at behavioural as well as organisational components within the hospital may gain additional importance, especially in regard of the growing financial pressure within the health sector. Trial registration DRKS The SEEGEN study is registered at the German Clinical Trial Register (DRKS) under the DRKS-ID DRKS00017249. Registered 08 October 2019, URL. https://www.drks.de/drks_web/navigate.do?navigationId=trial.HTML & TRIAL_ID=DRKS00017249 .
    Type of Medium: Online Resource
    ISSN: 1471-2458
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2019
    detail.hit.zdb_id: 2041338-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 602-602
    Abstract: Introduction Drug resistant cells represent a major threat for tumor patients as they might induce relapse and severely decrease disease outcome. Relapse represents a major drawback in patients with acute lymphoblastic leukemia (ALL), the single most frequent malignancy in children. Novel treatment options are intensively desired to remove drug resistant cells, which often additionally display dormancy. Aim We aimed at unraveling basic mechanisms determining drug resistance and dormancy, as basis for developing novel treatment strategies to prevent relapse. Methods Using cutting edge in vivo technology, we performed genetic engineering in the individualized xenograft mouse model of ALL. Primary patients' ALL cells were amplified in mice to generate patient-derived xenograft (PDX) cells. ALL PDX cells were lentivirally transduced to express transgenes. Recombinant luciferase allowed highly sensitive and reliable follow-up of leukemia growth and treatment. Recombinant surface markers enabled an unbiased approach to reliably and effectively enriching minute numbers of PDX cells from mouse bone marrow. Two independent, complementary innovative preclinical in vivo mouse models were established.In the first model, proliferation sensitive dyes allowed identifying and enriching in vivo long-term dormant PDX ALL cells.In the second model, the clinically highly relevant and challenging situation of MRD was mimicked in mice. PDX ALL cells were grown to advanced leukemia stages of above 30 % human blasts in bone marrow, when systemic chemotherapy with conventional cytotoxic drugs was initiated for prolonged periods of time, similar as applied in ALL patients. Chemotherapy reduced advanced leukemia down to 0,1 % or 10-3 leukemia cells in bone marrow, resembling not only complete morphologic remission, but even molecular remission. This novel preclinical model allows for the first time to characterize patients' dormant and MRD cells in detail including functional in vivo assays. Results Using our innovative preclinical model of dormancy, we identified a novel, distinct, rare subpopulation of PDX ALL cells that displayed long term dormancy in vivo. Long-term dormant cells showed significant resistance against drug treatment in vivo, as therapy nearly exclusively targeted proliferating cells. Dormant cells showed stem cell behavior as they initiated leukemia upon re-transplantation into further recipient mice. Long-term dormant cells thus combined the three challenging characteristics of relapse-inducing cells dormancy, drug resistance and stemness with re-growth upon withdrawal of treatment pressure. Using our second novel preclinical model, we isolated a pure, vivid fraction of rare MRD cells. These cells showed drug resistance in vivo and stemness features. We used single cell RNA sequencing to compare the transcriptomes of dormant and MRD populations and found that they were highly similar. Both populations had further similarities with primary high-risk ALL cells and dormant sub-fractions in patients' leukemia samples. Of high relevance for future treatment strategies, both, dormancy and drug resistance revealed transient characteristics in PDX ALL cells. When PDX long-term dormant ALL cells were distracted from their in vivo environment, they started proliferating similarly as their previously highly proliferative counterparts. When in vivo drug resistant PDX ALL cells were retrieved from murine bone marrow, they showed similar drug sensitivity in vitro as their sensitive counterparts. Summary/Conclusion Thus, both in vivo dormancy and drug resistance represent reversible characteristics in ALL cells which might result from the localization of ALL cells in the bone marrow niche. Dissolving ALL cells from their in vivo environment might sensitize them towards treatment. Addressing and inhibiting the interaction between ALL cells and their bone marrow niche might represent an attractive future therapeutic strategy to prevent ALL relapse. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 456-456
    Abstract: Acute lymphoblastic leukemia (ALL) is known to consist of several clones that might have different chromosomal, genetic or epigenetic aberrations. However, little is known about functional diversity in these different clones. In some patients, cells cannot be eradicated by standard therapy regimens, and aggressive or otherwise unfavorable clones might survive, eventually resulting in relapse and a poor prognosis of the patients. Here, we asked whether genetically distinct clones of ALL from a single patient would show a functionally distinct response towards drug treatment in vivo. As technical approach, we genetically engineered primary patients' ALL cells growing in immuno-compromized NSG mice as patient derived xenograft (PDX) cells by lentiviral transduction. ALL PDX cells were red-green-blue (RGB) color marked in order to discriminate several differently colored cell populations in the same mouse in functional in vivo experiments. ALL PDX cells further expressed luciferase for bioluminescence in vivo imaging (BLI) for sensitive and reliable monitoring of disease burden. Limiting dilution transplantation of RGB marked PDX cells transplanted into groups of mice allowed generating individually marked single cell clones which were discriminated by flow cytometry. Populations expressing a distinct color were sorted and analyzed by ligation mediated PCR to verify distinct integration of lentiviral inserts to prove single cell clone (SCC) origin of the population. In sum, eight distinct SCCs could be generated and were used for functional and -OMICs approaches. Targeted resequencing of the eight SCCs and the bulk cells revealed that all samples had mutations in CSMD1 and HERC1 with variant allele frequencies (VAF) of 0.5, indicating that these mutations represent the founding clone. However, we also found mutations that were only present in single samples: FAT1 and STAG2 mutations were found in SCC 3, whereas CSMD1 and USP6 mutations were found in SCC 6. Whole exome sequencing revealed SCC specific patterns, identifying SCC 6 being the clone furthest away from the bulk population. As the patient showed a high hyperdiploidy (+6,+13,+14,+17,+18,+21,+22,+X), we tested SCC and bulk cells by fluorescence in situ hybridization (FISH) and found that both the bulk sample and the SCCs consisted mainly of cells harboring three X chromosomes and to a minor proportion (between 2% and 20%) of cells harboring two X chromosomes. Only SCC 6 consisted exclusively of cells harboring two X chromosomes. Additionally, this SCC showed a distinct DNA-methylation pattern analyzed by 450K arrays (illumina). To analyze if the chromosomal, genetic and epigenetic differences also resulted in functional diversity, we first performed a competitive transplantation assay, injecting a mixture of five SCCs in the same ratio (20% each) into single mice. After 42 days when overt leukemia had established in the mice, cells were re-isolated and proportion of SCCs reanalyzed according to their specific color. Interestingly, SCC 5 (25%) and 7 (36%) had a clear growth advantage over SCCs 1 (14%), 6 (13%) and 8 (12%). The same pattern could be overserved if only SCC 5 (50% in, 92% out) and SCC 6 (50% in, 8% out) were transplanted. Next, response towards chemotherapeutic drugs was assessed. In vitro, SCC 6 was much more resistant towards the glucocorticoids prednisolon and dexamethasone (Dexa) compared to all other SCCs and bulk cells. Cells of SCC 5 and SCC 6 were mixed in equal amounts and transplanted into mice. Four days after transplantation, mice were randomized and treated with PBS or Dexa (2 or 8 mg/kg i.p., 5 days a week, 5 weeks). BLI showed a clear response towards therapy of the entire tumor. After 61 days, control treated mice showed again an outgrowth of SCC 5 (83% vs. 17% SCC 6), while Dexa treated animals showed the opposite pattern (Dexa 2 mg/kg: SCC 6 35%; Dexa 8 mg/kg: SCC 6 59%) indicating that SCC 6 was more resistant towards Dexa treatment in vivo. Taken together, our results clearly show that within a single ALL patient, genetically and functionally distinct subpopulations exist. Combining PDX model with genetic marking of the cells enables us to in-depth analyze SCCs of a single patient sample and eventually identify adverse prognostic markers. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2630-2630
    Abstract: Treatment-resistant cells determine prognosis and outcome of cancer patients as they induce relapse with poor outcome. Novel therapeutic options are urgently needed to eradicate chemo-resistant tumor cells. Towards this aim, a deep understanding is required on mechanisms determining treatment-resistance in vivo and the ability to induce relapse. Here, we aimed to identify and characterize the challenging rare subpopulation of drug resistant cells which survive in vivo chemo-therapy and are able to induce relapse. As technical approach, we used the individualized mouse model of acute lymphoblastic leukemia (ALL) and amplified primary tumor cells in mice to generate patient-derived xenograft (PDX) cells. Upon genetic engineering by lentiviral transduction, PDX ALL cells expressed the three transgenes NGFR, a red fluorochrome and luciferase. While recombinant luciferase was used for in vivo imaging, a combined MACS/FACS procedure based on the expression of the transgenes enabled enriching PDX ALL cells from murine bone marrow by a factor above 10,000. Staining of PDX cells with CFSE was used to discriminate between highly proliferative and dormant tumor cells in vivo. We treated mice harboring triple-transgenic, CFSE labeled PDX ALL cells with conventional chemotherapy; while in vivo treatment decreased the number of highly proliferative cells by more than 1 order of magnitude, the amount of dormant cells remained completely unchanged. Isolated, drug resistant cells revealed leukemia propagating potential and induced leukemia upon transplantation into next generation mice. Thus and using dormancy as an anchor, we could identify, isolate and enrich a subpopulation of treatment-resistant PDX ALL cells which might mimic relapse-inducing cells at minimal residual disease in patients. We next aimed at characterizing the expression profile of these cells and were able to isolate single cells out of the low number of dormant cells to perform single cell RNA sequencing. Dormant, drug-resistant cells showed increased expression of several adhesion molecules suggesting an increased dependence on the bone marrow environment. Upon using gene set enrichment analyses, drug-resistant cells showed a highly similar expression profile to primary high risk leukemia subpopulations such as primary high risk ALL cells, the subpopulation of CD34 positive CML cells and leukemic or benign hematopoietic stem cells. Taken together, treatment-resistant PDX ALL stem cells isolated and enriched from mice showed increased expression of adhesion molecules and resemble primary tumor cells of high risk subpopulations. These cells represent valuable tools to increase our understanding of mechanisms in minimal residual disease and relapse in patients. Our model will help to develop novel therapies which eliminate treatment resistant cells, prevent disease relapse and increase the prognosis of patients with ALL. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cellular and Molecular Life Sciences, Springer Science and Business Media LLC, Vol. 75, No. 24 ( 2018-12), p. 4539-4555
    Type of Medium: Online Resource
    ISSN: 1420-682X , 1420-9071
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 1458497-9
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 11, No. 1 ( 2020-12-17)
    Abstract: A Correction to this paper has been published: https://doi.org/10.1038/s41467-020-20453-0
    Type of Medium: Online Resource
    ISSN: 2041-1723
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2553671-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 391-391
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) has frequent alterations in few genes (KRAS, CDKN2A/TP53/SMAD4) and extensive heterogeneity of cancer drivers beyond. The expectation that mutational landscapes of rare drivers could explain phenotypic diversity has -with few exceptions- not come true. Likewise, PDAC metastasis is not understood, and comparisons of primary/metastasis pairs did not find recurrently mutated “metastasis genes”. Here we show that key aspects of PDAC biology are defined by gene-dosage variation of PDAC signature genes, evolving along distinct evolutionary routes. We found increased gene dosage of the initiating KRAS mutation (KRASMUT-iGD) in human PDAC precursors. Mouse models revealed the importance of KrasMUT-iGD for both, early progression and metastasis, rationalizing the high frequency of PDAC dissemination at diagnosis. To overcome limitations posed to gene dosage studies by PDAC´s stroma-richness, we developed murine cell culture resources comprising 135 primaries/metastases. Integrative analyses of their genomes, transcriptomes and tumor phenotypes, combined with human studies and functional analyses revealed a series of additional KrasMUT-dosage effects: different KrasMUT-levels define distinct cellular morphologies, histopathologies and clinical outcomes, with highest KrasMUT-expression underlying the most aggressive undifferentiated phenotypes. We also observed KrasMUT-dosage-associated cellular plasticity, including epithelial-to-mesenchymal transition. Mechanistically, oncogenic dosage-variation is linked to distinct evolutionary routes, characterized by defined types/states of tumor-suppressor alterations: Phylogenetic tracking studies revealed convergent evolution of KrasMUT-iGD-gains, with dependence on prior homozygous Cdkn2a- or Trp53-loss. By contrast, in Cdkn2aHET cancers, amplifications of known and novel oncogenes (Myc, Yap1, Nfkb2) collaborate with KrasMUT-HET to drive progression, yet with lower metastatic potential. These results also reveal oncogene-selective/context-dependent Cdkn2a-haploinsufficiency, for which Tgfβ pathway alterations provide permissiveness. Our study uncovers universal principles underlying PDAC biology and phenotypic diversification. It describes evolutionary trajectories, identifies their genetic hallmarks and shows how oncogenic dosage-variation is differentially licensed along individual routes to control critical disease characteristics, including early progression, histopathology, metastasis, cellular plasticity and clinical aggressiveness. Citation Format: Sebastian Mueller, Thomas Engleitner, Roman Maresch, Magdalena Zukowska, Sebastian Lange, Thorsten Kaltenbacher, Björn Konukiewitz, Rupert Öllinger, Maximilian Zwiebel, Alex Strong, Hsi-Yu Yen, Ruby Banerjee, Sandra Louzada, Beiyuan Fu, Barbara Seidler, Juliana Götzfried, Kathleen Schuck, Zonera Hassan, Nina Schönhuber, Sabine Klein, Christian Veltkamp, Mathias Friedrich, Lena Rad, Maxim Barenboim, Christoph Ziegenhain, Julia Hess, Oliver M. Dovey, Stefan Eser, Swati Parekh, Fernando Constantino-Casas, Jorge de la Rosa, Marta I. Sierra, Mario Fraga, Julia Mayerle, Günter Klöppel, Roland M. Schmid, Juan Cadiñanos, Pentao Liu, George Vassiliou, Wilko Weichert, Katja Steiger, Wolfgang Enard, Fengtang Yang, Kristian Unger, Günter Schneider, Ignacio Varela, Allan Bradley, Dieter Saur, Roland Rad. Evolutionary trajectories and KRAS gene dosage define pancreatic cancer phenotypes [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 391.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages