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
    In: Cell Reports, Elsevier BV, Vol. 42, No. 10 ( 2023-10), p. 113017-
    Type of Medium: Online Resource
    ISSN: 2211-1247
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 2649101-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2022
    In:  Blood Vol. 140, No. Supplement 1 ( 2022-11-15), p. 606-607
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 606-607
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Science, American Association for the Advancement of Science (AAAS), Vol. 380, No. 6640 ( 2023-04-07), p. 93-101
    Abstract: Most drugs act on proteins and engage cellular pathways regulated by protein posttranslational modifications (PTMs), such as phosphorylation, acetylation, or ubiquitinylation, to exert their therapeutic effects. Because polypharmacology is common, it is important to characterize drugs on a proteome-wide scale to understand all their mechanisms of action. RATIONALE There is a lack of dose- and time-dependent drug characterization at the level of proteins and PTMs, arguably the most important characteristics of drug action in any biological context. To address this gap, this study presents a quantitative proteomic approach called decryptM, which is able to assess drug target and pathway engagement as well as cellular mechanism of action by measuring thousands of PTM responses in a dose- and time-resolved fashion. RESULTS DecryptM profiling of 31 cancer drugs in 13 human cancer cell line models resulted in 1.8 million dose-response curves, including 47,502 regulated phosphopeptides, 7316 ubiquitinylated peptides, and 546 regulated acetylated peptides, all of which can be explored in ProteomicsDB ( www.proteomicsdb.org/decryptM ). The observed close coherence between drug-target affinity and drug-PTM modulation potency enabled placing functionally uncharacterized PTM sites into known pathways and thus decrypting them on the grounds of guilt by association. Examples include previously uncharacterized phosphorylation sites linking chemotherapeutic drugs to the DNA damage response, regulated phosphorylation sites indicating breakdown of oncogenic signaling in response to phosphatase and kinase inhibitors, or activation of the unfolded protein response upon proteasome inhibition. Similarly, decryptM profiles enabled the identification of previously unknown substrates of kinases as well as lysine acetyltransferases and deacetylases. Each drug appeared to leave a cell line–specific decryptM signature that may constitute a pharmacodynamic marker for target and pathway engagement, identify points of conversion of signaling axes, or distinguish closely related compounds. DecryptM analysis of therapeutic anti-HER2 antibodies revealed differences in their mechanisms of action. Whereas pertuzumab cuts off the HER3–mitogen-activated protein kinase (MAPK) and HER3-PI3K/AKT signaling axis in breast cancer cells, trastuzumab had no effect on any of the phosphoproteomes investigated. By contrast, the anti-CD20 antibody rituximab massively activates signaling in B cells. The collective decryptM and functional evidence supports a model in which rituximab binds to CD20, located in lipid rafts alongside the B cell receptor complex, and leads to strong activation of the MAPK and calcineurin–nuclear factor of activated T cells (NFAT) axis, tipping the signaling balance toward apoptotic cell death. CONCLUSION The examples presented in this study illustrate the potential of decryptM to characterize drugs’ mechanisms of action, generate drug-specific PTM signatures, study resistance mechanisms, and place drug-regulated PTM sites of unclear significance into a functional context. The workflow developed supports decryptM profiling at scale and should be extendable to any molecule that modulates cellular activity by affecting PTMs or protein expression. This may include G protein–coupled receptor (GPCR) ligands, cytokines, chemokines, cofactors, metabolites, biologics, peptides, and hormones among many other factors. In the future, decryptM profiles may also serve to monitor and potentially predict drug responses in vivo. Looking further, we envision that matching decryptM profiles of cancer drugs with PTM profiles of cancer patients will become important for evidence-based treatment recommendations by molecular tumor boards. Schematic representation of the decryptM approach. Following the dose-dependent treatment of cancer cells with molecularly targeted drugs, quantitative mass spectrometry can record the dose-response characteristics of thousands of posttranslationally modified peptides in parallel. Such drug profiles can be used to place previously uncharacterized modification sites into a functional context and elucidate the cellular mechanism of action of a given drug. EC 50 , half-maximal effective concentration; Ac, acetylation; GG, ubiquitinylation; P, phosphorylation.
    Type of Medium: Online Resource
    ISSN: 0036-8075 , 1095-9203
    RVK:
    RVK:
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2023
    detail.hit.zdb_id: 128410-1
    detail.hit.zdb_id: 2066996-3
    detail.hit.zdb_id: 2060783-0
    SSG: 11
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Experimental Medicine, Rockefeller University Press, Vol. 218, No. 2 ( 2021-02-01)
    Abstract: Clinical evidence suggests alterations in receptor activator of NF-κB (RANK) signaling are key contributors to B cell autoimmunity and malignancy, but the pathophysiological consequences of aberrant B cell–intrinsic RANK signaling remain unknown. We generated mice that express a human lymphoma–derived, hyperactive RANKK240E variant in B lymphocytes in vivo. Forced RANK signaling disrupted B cell tolerance and induced a fully penetrant systemic lupus erythematosus–like disease in addition to the development of chronic lymphocytic leukemia (CLL). Importantly, RANKK240E transgenic CLL cells as well as CLL cells of independent murine and of human origin depend on microenvironmental RANK ligand (RANKL) for tumor cell survival. Consequently, inhibition of the RANKL–RANK axis with anti-RANKL antibodies killed murine and human CLL cells in vitro and in vivo. These results establish pathological B cell–intrinsic RANK signaling as a potential driver of autoimmunity and B cell malignancy, and they suggest the exploitation of clinically available anti-RANKL compounds for CLL treatment.
    Type of Medium: Online Resource
    ISSN: 0022-1007 , 1540-9538
    RVK:
    Language: English
    Publisher: Rockefeller University Press
    Publication Date: 2021
    detail.hit.zdb_id: 1477240-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Proceedings of the National Academy of Sciences ; 2018
    In:  Proceedings of the National Academy of Sciences Vol. 115, No. 12 ( 2018-03-20), p. 3120-3125
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 115, No. 12 ( 2018-03-20), p. 3120-3125
    Abstract: The transcription factor Foxp1 is critical for early B cell development. Despite frequent deregulation of Foxp1 in B cell lymphoma, the physiological functions of Foxp1 in mature B cells remain unknown. Here, we used conditional gene targeting in the B cell lineage and report that Foxp1 disruption in developing and mature B cells results in reduced numbers and frequencies of follicular and B-1 B cells and in impaired antibody production upon T cell-independent immunization in vivo. Moreover, Foxp1 -deficient B cells are impaired in survival even though they exhibit an increased capacity to proliferate. Transcriptional analysis identified defective expression of the prosurvival Bcl-2 family gene Bcl2l1 encoding Bcl-xl in Foxp1-deficient B cells, and we identified Foxp1 binding in the regulatory region of Bcl2l1 . Transgenic overexpression of Bcl2 rescued the survival defect in Foxp1-deficient mature B cells in vivo and restored peripheral B cell numbers. Thus, our results identify Foxp1 as a physiological regulator of mature B cell survival mediated in part via the control of Bcl-xl expression and imply that this pathway might contribute to the pathogenic function of aberrant Foxp1 expression in lymphoma.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2018
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 12, No. 1 ( 2021-06-10)
    Abstract: Current therapeutic approaches for chronic lymphocytic leukemia (CLL) focus on the suppression of oncogenic kinase signaling. Here, we test the hypothesis that targeted hyperactivation of the phosphatidylinositol-3-phosphate/AKT (PI3K/AKT)-signaling pathway may be leveraged to trigger CLL cell death. Though counterintuitive, our data show that genetic hyperactivation of PI3K/AKT-signaling or blocking the activity of the inhibitory phosphatase SH2-containing-inositol-5′-phosphatase-1 (SHIP1) induces acute cell death in CLL cells. Our mechanistic studies reveal that increased AKT activity upon inhibition of SHIP1 leads to increased mitochondrial respiration and causes excessive accumulation of reactive oxygen species (ROS), resulting in cell death in CLL with immunogenic features. Our results demonstrate that CLL cells critically depend on mechanisms to fine-tune PI3K/AKT activity, allowing sustained proliferation and survival but avoid ROS-induced cell death and suggest transient SHIP1-inhibition as an unexpectedly promising concept for CLL therapy.
    Type of Medium: Online Resource
    ISSN: 2041-1723
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2553671-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Hematology ; 2018
    In:  Blood Vol. 132, No. Supplement 1 ( 2018-11-29), p. 894-894
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 894-894
    Abstract: Chronic lymphocytic leukemia (CLL) is one of the most common B cell malignancies in the Western world. Malignant B cells are blocked from differentiating into immunoglobulin producing-plasma cells and clonally accumulate in the spleen, lymph nodes, bone marrow and peripheral blood. CLL is characterized by immunosuppression throughout all disease stages, which is mediated by increased numbers of myeloid-derived suppressor cells (MDSCs), regulatory T cells (Jitschin and Braun et al., Blood 2014) and direct inhibitory effects of the malignant CLL cells on T cells (Christopoulos etal., Blood 2011). Over the past decade, significant improvement in understanding the pathogenesis of CLL has highlighted the importance of active B cell receptor (BCR) signaling. This has revealed promising targeted treatment options, including the small molecule inhibitors targeting the phosphatidylinositol-3-kinase (PI3K) signaling pathway. Idelalisib and Duvelisib are under clinical investigation for CLL treatment, however, treatment-related toxicities are limiting their application and none of these approaches are curative, highlighting the importance of functional anti-tumor immune responses in CLL for prolonged treatment efficacy. Here, we are testing a novel approach that aims to selectively target CLL B cells and simultaneously restore an appropriate immune cell function. The phosphatase SH2-domain-containing inositol 5ʹ-phosphatase 1 (SHIP1) serves as negative feedback molecule and downregulates PI3K signaling in B cells by dephosphorylating the 5`phosphate of Phosphatidylinositol (3,4,5)-trisphosphate. We hypothesize that CLL cells rely on such negative regulators for optimal survival and can only tolerate a maximum signaling level. We are therefore testing whether SHIP1 inhibition induces hypersignaling and thereby CLL cell death. Furthermore, we are investigating whether SHIP1 inhibition simultanously stimulates immune responses, as it has been shown to induce expansion of murine hematopoietic and mesenchymal stem cell compartments (Brooks et al., Stem cells 2014). 3α-Aminocholestane (3AC) is a small molecule inhibitor of SHIP1 and can be used for pharmacological inhibition. First, we investigated the expression and phosphorylation levels of SHIP1 in CLL. We found SHIP1 to be expressed at various levels in CLL peripheral blood and strongly phosphorylated compared to age-matched healthy donors. Besides, SHIP1 transcription is upregulated in lymph nodes as compared to peripheral blood, which is in line with the assumption of increased BCR signaling in secondary lymphoid organs. We then set out to investigate the consequences of SHIP1 phosphatase inhibition. Similarly, to recent findings in acute lymphoblastic leukemia (Chen et al., Nature 2015), pharmacological inhibition of SHIP1 lead to rapid cell death of CLL cells. We further investigated the mode of cell death and observed specific features of apoptosis, namely caspase 3/7 activation and phosphatidylserine exposure on the outer cell membrane. This has been tested on primary CLL patient samples and T cell leukemia/lymphoma 1 (TCL1)-driven murine CLL cells and was not observed or significantly less pronounced in other lymphoma cell lines or healthy primary B cells. To confirm the specificity of the observed effects, we genetically activated AKT with a GFP reporter in the TCL1-driven mouse model in vivo and in vitro. By tracking GFP-expressing CLL cells, we observed an initial expansion followed by rapid cell death in vitro. When we induced AKT activation in vivo, GFP+ CLL cells were not detectable in the peripheral blood, total CLL count declined upon induction and we found decreased tumor burden in the secondary lymphoid organs, particularly in the lymph nodes. In addition to the direct effects on CLL cells, we sought to investigate the impact of SHIP1 inhibition on other immune cell functions. We observed that SHIP1 inhibition lowers the activity threshold of T cells: When we stimulated a reporter cell line with suboptimal doses of anti-CD3, 3AC treatment significantly enhanced the response rate. Therefore, we propose SHIP1 as a novel interesting target in CLL. In contrast to kinase inhibition and downregulation of the BCR signaling strength, phosphatase inhibition and BCR signaling overaction provides an attractive new treatment strategy for elimination of malignant CLL cells and stimulation of immune responses. 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 ...
  • 8
    In: Molecular Cell, Elsevier BV, Vol. 81, No. 10 ( 2021-05), p. 2094-2111.e9
    Type of Medium: Online Resource
    ISSN: 1097-2765
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 2001948-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Society of Hematology ; 2018
    In:  Blood Vol. 132, No. Supplement 1 ( 2018-11-29), p. 2857-2857
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 2857-2857
    Abstract: Chronic lymphocytic leukemia (CLL) is characterized by an expansion of monoclonal, mature B cells that carry auto/polyreactive B cell receptors (BCRs). It is one of the most prevalent B cell malignancies in Western countries. Over the past decade, extensive research has led to significant improvement in understanding its pathogenesis, particularly in regard to the BCR signaling pathway. This has led to the development of novel and promising treatment options, however, none of these approaches are curative and relapses are challenging to treat. Until now allogeneic hematopoietic stem cell transplantation remains the only treatment option for durable remission in poor-risk disease. The therapeutic difficulties result from the heterogeneity, the advanced age of the patients, chemoresistance and relapse of the disease. Here, we test a novel strategy for simultaneously targeting the malignant cells and reverting the CLL-suppressed immune response. Stimulation of the antigen receptors of immune cells readily induces proliferation, differentiation and functional activation. However, strong binding of antigen can also induce negative selection in lymphocytes. CLL B cells constitutively signal through their BCR and thus we set out to investigate whether they rely on the counterbalancing, negative regulation of specific downstream signaling pathways, in particular the mitogen-activated protein kinase (MAPK) pathway. The dual specificity phosphatases 1 and 6 (DUSP1 and DUSP6) dephosphorylate extracellular-signal regulated kinase 1/2 (ERK1/2) and thereby limit ERK1/2 activation. These molecules are frequently downregulated in solid tumors (Khor et al., Int. J. Med.Sci 2013; Okudela et al., Am. J. Pathol. 2009). We therefore set out to analyze the expression levels of DUSP1 and DUSP6 in CLL and found them readily expressed at various levels, comparable to normal B cells. To determine the functional relevance of DUSP1 and DUSP6, we blocked their phosphatase function using the small molecule inhibitor BCI. By treatment with BCI, we induced hyperactivation of the MAPK signaling cascade followed by cell death of the CLL cells. Interestingly, the induction of cell death is specific for CLL cells and does not occur to the same extent in other malignant B cell lymphoma cells or healthy donor-derived B cells. This deleterious effect of BCI was evident in primary patient-derived CLL cells as well as in the CLL-like cell lines MEC-1 and EHEB and in CLL cells derived from the T cell leukemia/lymphoma 1 (TCL1)-driven mouse model. To further investigate the downstream signaling event upon BCI treatment, we conducted a global phosphoproteome analysis. After treatment of primary CLL cells with BCI, the most significant alterations were within the BCR signaling pathway, including hyperphosphorylation of ERK1/2 and followed by a rapid induction of a DNA damage response. These results were validated by immunoblot analysis of human and murine CLL cells and were not detected in BCI-resistant cell lines. Beside the direct effects of BCI on the CLL cells we set out to investigate effects on other immune cells, directly by BCI and secondary via CLL cells treated with BCI. Indeed, we observed changes in immune cell compartment: BCI-treated CLL patient-derived peripheral blood mononuclear cells (PBMCs) resulted in selective enrichment of cytotoxic T cells. Furthermore, BCI treatment of CLL cells fed with Ovalbumin, in co-culture with or without BMDCs and OT-I cytotoxic CD8 T cells (specifically recognizing the SINFEKL peptide), resulted in the induction of immunogenic cell death of CLL cells. This was evidenced by enhanced antigen-specific T cell proliferation and release of the high mobility group box 1 protein (HMGB1). To investigate indirect effects of BCI, we treated CLL cells with BCI at sublethal doses, then washed the cells and co-cultured these primary CLL cells or CLL cell lines with healthy donor-derived PBMCs. The frequencies of CLL-induced myeloid-derived suppressor cells (MDSCs) as well as regulatory T cells (Tregs) were reduced after co-cultivation of PBMCs with BCI-pretreated CLL cells. Taken together, our data indicate that negative feedback inhibition reduces CLL content by induction of immunogenic cell death and activates immune cells to target the CLL-induced dysfunction of the immune system. We therefore propose that inhibition of DUSP1/6 is a promising therapeutic approach for CLL. 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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 9840-9841
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    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...