GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    Elsevier BV ; 2002
    In:  The Journal of Molecular Diagnostics Vol. 4, No. 2 ( 2002-5), p. 90-96
    In: The Journal of Molecular Diagnostics, Elsevier BV, Vol. 4, No. 2 ( 2002-5), p. 90-96
    Type of Medium: Online Resource
    ISSN: 1525-1578
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2002
    detail.hit.zdb_id: 2032654-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Annual Reviews ; 2018
    In:  Annual Review of Pathology: Mechanisms of Disease Vol. 13, No. 1 ( 2018-01-24), p. 193-217
    In: Annual Review of Pathology: Mechanisms of Disease, Annual Reviews, Vol. 13, No. 1 ( 2018-01-24), p. 193-217
    Abstract: Lymphomas represent clonal proliferations of lymphocytes that are broadly classified based upon their maturity (peripheral or mature versus precursor) and lineage (B cell, T cell, and natural killer cell). Insights into the pathogenetic mechanisms involved in lymphoma impact the classification of lymphoma and have significant implications for the diagnosis and clinical management of patients. Serial scientific and technologic advances over the last 30 years in immunology, cytogenetics, molecular biology, gene expression profiling, mass spectrometry–based proteomics, and, more recently, next-generation sequencing have contributed to greatly enhance our understanding of the pathogenetic mechanisms in lymphoma. Novel and emerging concepts that challenge our previously accepted paradigms about lymphoma biology and how these impact diagnosis, molecular testing, disease monitoring, drug development, and personalized and precision medicine for lymphoma are discussed.
    Type of Medium: Online Resource
    ISSN: 1553-4006 , 1553-4014
    URL: Issue
    Language: English
    Publisher: Annual Reviews
    Publication Date: 2018
    detail.hit.zdb_id: 2217576-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: The Journal of Molecular Diagnostics, Elsevier BV, Vol. 25, No. 2 ( 2023-02), p. 87-93
    Type of Medium: Online Resource
    ISSN: 1525-1578
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 2032654-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3925-3925
    Abstract: Abstract 3925 Poster Board III-861 Introduction Multiple myeloma oncogene 1/interferon regulatory factor 4 (MUM1/IRF4) is a 52-kDa transcriptional activator protein which plays an important role in interferon-stimulated response element (ISRE)-regulated signal transduction mechanisms important in lymphoid cell development and differentiation. MUM1/IRF4 is also critical for the generation of immunoglobulin-secreting plasma cells. MUM1/IRF4 is expressed in the activated B-cell (ABC)-like subset of diffuse large B-cell lymphomas (DLBCL) and is targeted by chromosomal translocations in a subset of multiple myelomas and peripheral T-cell lymphomas. Despite its importance in lymphocyte and lymphoma biology, and its addiction demonstrated in multiple myelomas, the proteomic network of MUM1/IRF4 interacting proteins has not been determined. Methods We determined the proteomic interaction networks of MUM1/IRF4 in B-cell and T-cell lymphoma contexts, by using a functional proteomic approach. Total cell lysates were prepared from MUM1/IRF4 expressing cell lines including OCI-LY10 ((ABC)-like DLBCL) and HH (mycosis fungoides derived-T-cell) and, as a negative control, from non-MUM1/IRF4 expressing SUDHL4 cell line. Immunoprecipitates of the MUM1/IRF4 expressing B- and T-cell line were compared to that of hyperimmune mouse immunoglobulin by 1-dimensional sodium dodecyl sulphate polyacrylamide gel electrophoresis. Coomassie-stained protein bands from both immunocomplexes were excised and analyzed by electrospray liquid chromatography tandem mass spectrometry (ESI-LC/MS/MS). Peptide sequences were identified by searching the MS/MS data against human IPI protein using database X!Tandem with k-score plug-in. Proteins found in the two control sets (SUDHL4 and IgG isotype) were manually subtracted from the proteins found in the experimental set. MS/MS results were validated using immunoprecipitation and Western blotting and some of the interacting proteins were further confirmed by reciprocal immunoprecipitation. Results A total of 163 and 94 proteins were identified from 12 Coomassie-stained bands which were unique to the MUM1/IRF4 immunocomplex in OCI-Ly10 and HH cell lines, respectively. Previously reported proteins in the MUM1/IRF4 signal pathway were identified including FK506 and TRAF family protein. More importantly, many proteins previously not associated with MUM1/IRF4 were identified. In common to both subsets were proteins such as ARHGDIA-involved in RhoA-mediated signaling. In the ABC-like B-cell (OCI-LY10) specific context, these included proteins known to be critical regulators of lymphocyte proliferation (RAB2A), motility, trafficking and cell adhesion. In the T-cell specific context, proteins known to play important roles in endocytosis (Reggies) and T-cell activation (GDI2, Guanine nucleotide exchange factors) were identified in the MUM1/IRF4 interactome. A subset of the proteins identified by MS were confirmed by western blotting and reciprocal immunoprecipitation. Conclusions Our studies reveal that although a minor subset of MUM1/IRF4 interacting proteins are common in B- and T-cells, MUM1/IRF4 exhibits distinct interaction partners dependent of specific cellular contexts. The diverse interaction networks implicate MUM1/IRF4 in previously undescribed functional roles in lymphocyte-specific subsets. Comprehensive elucidation of the protein-protein interaction networks in the specific cellular contexts will provide opportunities for exploitation of the knowledge for design of rational interventions targeting the critical nodes and modules in MUM1/IRF4-deregulation in B- and T-cell malignancies. 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: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Society of Hematology ; 2009
    In:  Blood Vol. 114, No. 22 ( 2009-11-20), p. 1277-1277
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 1277-1277
    Abstract: Abstract 1277 Poster Board I-299 Mixed lineage leukemia (MLL) is a histone H3 lysine 4 methyltransferase that is required to maintain a normal hematopoietic stem cell compartment. MLL functions to maintain expression of HOX genes as well as the HOX co-factor MEIS1, which play significant roles in regulating hematopoiesis. MLL is involved in chromosomal translocations with up to sixty different partners in both acute myeloid leukemia (AML) and acute lymphoid leukemia (ALL). HOXA9 and MEIS1, are directly regulated by MLL fusion proteins and are crucial for MLL fusion protein mediated transformation. The deregulated expression of target genes in AML is dependent on specific protein-protein interactions and functional domains of MLL. For example, the tumor suppressor Menin bridges LEDGF to the extreme N-terminus of MLL and both of these interactions are necessary for transformation. Furthermore, a DNA methyltransferase homology region (CxxC domain) of MLL is essential for binding to non-methylated CpG islands and MLL-fusion protein oncogenesis. We have found that sequences downstream of the CxxC domain, termed the RD2 region, that interact with the Polymerase Associated Factor (PAF) complex are also required for MLL fusion protein mediated transformation. The PAF complex interacts with RNA polymerase II and is required for H2B mono-ubiquitination and subsequent histone H3K4 and H3K79 methylation. Together the PAF complex has been shown to be involved in transcriptional initiation, elongation and termination. Interaction of MLL with the PAF complex is mediated through direct contacts with two subunits: Ctr9 and PAF1. The PAF complex synergizes with MLL-AF9 to augment transcriptional activation of the Hoxa9 promoter. Furthermore, MLL fusion proteins recruit high levels of the PAF complex to the Hoxa9 promoter. Importantly, deletions of the MLL RD2 region that abolish interactions with the PAF complex eliminate MLL-AF9 mediated transformation of mouse bone marrow cells. Transcription of PAF components is dramatically downregulated during differentiation of hematopoietic cells, consistent with recent data showing a requirement for the PAF complex to maintain an embryonic stem cell phenotype. Knock down and transplantation experiments are underway to further define how the PAF complex regulates normal MLL function and cooperates with MLL fusion proteins to promote leukemogenesis. 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: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 780-780
    Abstract: Background: The genetic basis and the genome-wide abnormalities underlying most forms of cancer are being comprehensively annotated in many human malignancies. However, the contribution of epigenetic aberrations, particularly the post-translational modifications (PTM) of histone tails has not been investigated. This is largely due to lack of approaches to comprehensively interrogate the status of the numerous PTMs that define histone marks which control gene expression and complex biologic processes such as cancer. Sézary Syndrome (SS) is an aggressive form of cutaneous T-cell lymphoma (CTCL) characterized by poor outcomes and complex genetic alterations frequently targeting epigenetic regulators and chromatin remodelers. CTCLs represent the first FDA-approved disease for treatment using histone deacetylase inhibitors (HDACi) such as romidepsin, but the direct consequences of this treatment on histone PTMs remain unknown. Here we define the histone PTM signatures of CTCL/SS by using a novel unbiased strategy leveraging tandem mass spectrometry (MS/MS)-based quantitative proteomics to study the comprehensive combinatorial histone PTM code in a cohort of primary SS samples, and 4 CTCL/SS cell lines in comparison with normal reactive CD4+ T-lymphocytes from healthy individuals. Methods: CD4+ T-cells were isolated using CD4 immunomagnetic beads from PBMC's of healthy volunteers (n=8) or patients with diagnosed SS (n=20). CD4+ cells were frozen and subsequently analyzed in batches. The CTCL/SS-derived cell lines (HH and MJ, HuT78 and H9) were also analyzed each with three biological replicates. Histones were acid extracted from isolated nuclei. Total bulk histones were propionylated and trypsinized prior to MS analysis. Nano-scale liquid chromatography followed by -tandem mass spectrometry (nano LC-MS/MS) data acquisition was performed in an Orbitrap FusionTM TribridTM MS in technical triplicates for each sample with a data-independent acquisition (DIA)-method using a 50 m/z quadrupole-isolation windows that steps across the 200-1500 m/z ranges. Data analysis was performed using EpiProfile for single and combinatorial histone PTM analysis and quantification. Orthogonal validation for selected modifications was performed using western blotting and single-cell mass cytometry by time of flight (CyTOF) analysis. Results: Our quantitative epiproteomic strategy interrogated the relative ratios of a total of 228 histone PTM combinations, and 20 histone variants in a well-characterized cohort of SS patient samples, four CTCL cell lines and a cohort of normal primary CD4+ T-lymphocytes isolated from healthy individuals. With this approach, we were able to identify and quantify 23 unique histone peptides on histone H4, 105 on histone H3, 80 on histone H2A, 12 on histone H2B and 28 on histone H1 across every sample. Pearson correlation and principal component analysis of overall histone PTMs profiles across all samples provided accurate discrimination. We found a distinct pattern of histone PTMs in both SS patient samples and CTCL cell lines that were strikingly different from CD4+ T-cells obtained from healthy individuals. Notably, differences between cell lines and primary patient samples are more marked than those within individual SS primary patient samples, implying that cell lines may poorly recapitulate some aspects of in vivo biology. Unsupervised hierarchical clustering of the primary patient samples, cell lines and the normal CD4+ T-cells performed based on abundances of the identified histone PTMs revealed disease-specific ubiquitous marks as well as disease-specific unique marks for SS. Among the analyzed histone PTMs, H3K27ac, H3.3K27ac, H4K8ac, H4K20ac, H3K4me3, H3K18me1, H3K79me3 and H4K20me3 were distinct between healthy and CTCL CD4+ T-cells. Selected differential marks such as H4K20me3 were orthogonally corroborated by Western blotting and CyTOF mass cytometry analysis. Conclusions: For the first time, we have defined the histone code of CTCL/SS using global mass spectrometry based quantitative proteomics with high specificity and sensitivity. The results of our MS-based epiproteomic profiling revealed disease-specific histone PTM signatures and opportunities to exploit tractable changes induced by HDACi treatment as clinically actionable molecular biomarkers and therapeutic targets. 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: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3533-3533
    Abstract: HOXA9 helps maintain the balance between hematopoietic stem/progenitor cell self-renewal and myeloid/lymphoid cell differentiation in the bone marrow (BM). HOXA9 encodes a homeodomain-containing transcription factor, which mediates its function in collaboration with other co-factors, e.g., MEIS1. Several genetic alterations observed in acute myeloid leukemia (AML) patients, e.g., chromosomal translocations involving the MLL gene, are associated with aberrant upregulation of HOXA9, thus disrupting the hematopoietic balance towards leukemogenesis. In a proteomic screen combining the use of affinity purification coupled to mass spectrometry analysis (AP-MS) and the yeast two-hybrid system (Y2H), we discovered that HOXA9 interacts physically with OGT, an O-linked N-acetyl glucosamine transferase. We have obtained multiple lines of evidence supporting the HOXA9-OGT interaction: i) Y2H; ii) identification of endogenous OGT by affinity purification using HOXA9 as a bait in both murine leukemic blasts and human THP1 AML cells; and iii) reciprocal protein pulldowns in HEK293T cells (Fig. 1). A domain mapping analysis revealed that the HOXA9 domain D62-135 is required for the OGT-HOXA9 interaction. We also demonstrated that the MEIS interaction motif of HOXA9 is GlcNAcylated by OGT. We used a colony forming assay to measure the clonogenic potential of the OGT-interaction defective allele of HOXA9, i.e., HOXA9(Δ62-135). We observed a higher number of colonies for HOXA9(Δ62-135) in comparison to WT HOXA9 (Fig. 2). In light of these results, we hypothesize that the HOXA9-OGT interaction has an inhibitory effect on HOXA9's ability to promote colony formation. This model is also corroborated by an experiment in which co-transduction of OGT with HOXA9 inhibits clonogenesis, further supporting the hypothesis that the OGT has a suppressive effect on HOXA9. By quantitative RT-PCR, we show that the level of expression of FLT3, a well-characterized target of HOXA9, is higher in HOXA9(Δ62-135)-transduced BM cells than in HOXA9-transduced ones. To identify genes modulated by HOXA9 in an OGT-dependent manner, we analyzed the transcriptome of HOXA9(Δ62-135)-transduced BM cells by expression profiling studies using next-generation sequencing (RNA-seq). We identified 1,083 and 551 genes that are further up- or further down-regulated with HOXA9(Δ62-135) in comparison to HOXA9. Increased chromatin binding of HOXA9(Δ62-135) at the loci of some of these genes was also confirmed by chromatin immunoprecipitation followed by qPCR (ChIP-qPCR). We note that pro-oncogenic genes such as FLT3, LCK and ERG, which have been previously characterized as being HOXA9-upregulated, demonstrate even higher levels of expression in cells transformed by HOXA9(Δ62-135). The most striking observation, though, arose from the integrative analysis of two RNA-seq datasets [HOXA9 and HOXA9(Δ62-135)-transformed BM cells] and two ChIP-seq datasets (HOXA9 and OGT ChIP-seq in BM cells). Remarkably, we observed that HOXA9-downregulated genes that are further downregulated in presence of HOXA9(Δ62-135) (N = 82) are specifically enriched for OGT-bound genes (N = 41 out of 82; P 〈 4 x 10-10). In contrast, such enrichment is not observed for HOXA9-upregulated genes (N = 22 OGT-bound genes out of 108 HOXA9/HOXA9(Δ62-135)-upregulated genes; no statistical enrichment). Thus, our transcriptome-wide analysis of the OGT-dependent regulation of HOXA9 gene targets highlights the critical importance of HOXA9 as a negative regulator of transcription, rather than a positive regulator. This observation is further validated by the analysis of gene expression profiles in human leukemia samples. Indeed, a larger than expected number of the human orthologs of the murine HOXA9-downregulated genes have a low level of expression in MLL translocation-driven AML patient samples compared normal samples. In comparison, such trend is not as prevalent for the human orthologs of the murine HOXA9-upregulated genes. In conclusion, our data support a model in which OGT inhibits HOXA9's ability to transform primary bone marrow cells, thus defining OGT as a potential tumor suppressor of HOXA9-driven AML. The biomedical relevance of the OGT-HOXA9 interaction to HOXA9-driven leukemogenesis is being investigated in vivo using a HOXA9-induced mouse model of AML. This project is supported by the WES Foundation and the American Society of Hematology. 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: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    Ovid Technologies (Wolters Kluwer Health) ; 2018
    In:  Current Opinion in Pulmonary Medicine Vol. 24, No. 3 ( 2018-05), p. 281-286
    In: Current Opinion in Pulmonary Medicine, Ovid Technologies (Wolters Kluwer Health), Vol. 24, No. 3 ( 2018-05), p. 281-286
    Type of Medium: Online Resource
    ISSN: 1070-5287
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2018
    detail.hit.zdb_id: 2026974-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Ovid Technologies (Wolters Kluwer Health) ; 2015
    In:  Current Opinion in Hematology Vol. 22, No. 4 ( 2015-07), p. 369-378
    In: Current Opinion in Hematology, Ovid Technologies (Wolters Kluwer Health), Vol. 22, No. 4 ( 2015-07), p. 369-378
    Type of Medium: Online Resource
    ISSN: 1065-6251
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2015
    detail.hit.zdb_id: 2026995-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: The American Journal of Pathology, Elsevier BV, Vol. 153, No. 5 ( 1998-11), p. 1541-1550
    Type of Medium: Online Resource
    ISSN: 0002-9440
    RVK:
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 1998
    detail.hit.zdb_id: 1480207-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...