GLORIA

GEOMAR Library Ocean Research Information Access

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
  • 1
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 7, No. 1 ( 2016-04-27)
    Kurzfassung: Common variants in 94 loci have been associated with breast cancer including 15 loci with genome-wide significant associations ( P 〈 5 × 10 −8 ) with oestrogen receptor (ER)-negative breast cancer and BRCA1 -associated breast cancer risk. In this study, to identify new ER-negative susceptibility loci, we performed a meta-analysis of 11 genome-wide association studies (GWAS) consisting of 4,939 ER-negative cases and 14,352 controls, combined with 7,333 ER-negative cases and 42,468 controls and 15,252 BRCA1 mutation carriers genotyped on the iCOGS array. We identify four previously unidentified loci including two loci at 13q22 near KLF5 , a 2p23.2 locus near WDR43 and a 2q33 locus near PPIL3 that display genome-wide significant associations with ER-negative breast cancer. In addition, 19 known breast cancer risk loci have genome-wide significant associations and 40 had moderate associations ( P 〈 0.05) with ER-negative disease. Using functional and eQTL studies we implicate TRMT61B and WDR43 at 2p23.2 and PPIL3 at 2q33 in ER-negative breast cancer aetiology. All ER-negative loci combined account for ∼11% of familial relative risk for ER-negative disease and may contribute to improved ER-negative and BRCA1 breast cancer risk prediction.
    Materialart: Online-Ressource
    ISSN: 2041-1723
    Sprache: Englisch
    Verlag: Springer Science and Business Media LLC
    Publikationsdatum: 2016
    ZDB Id: 2553671-0
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: NeuroImage: Clinical, Elsevier BV, Vol. 29 ( 2021), p. 102533-
    Materialart: Online-Ressource
    ISSN: 2213-1582
    Sprache: Englisch
    Verlag: Elsevier BV
    Publikationsdatum: 2021
    ZDB Id: 2701571-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    In: British Journal of Haematology, Wiley, Vol. 169, No. 6 ( 2015-06), p. 824-833
    Kurzfassung: Ruxolitinib ( INCB 018424) is the first JAK 1/ JAK 2 inhibitor approved for treatment of myelofibrosis. JAK / STAT ‐signalling is known to be involved in the regulation of CD 4 + T cells, which critically orchestrate inflammatory responses. To better understand how ruxolitinib modulates CD 4 + T cell responses, we undertook an in‐depth analysis of CD 4 + T cell function upon ruxolitinib exposure. We observed a decrease in total CD 3 + cells after 3 weeks of ruxolitinib treatment in patients with myeloproliferative neoplasms. Moreover, we found that the number of regulatory T cells (Tregs), pro‐inflammatory T‐helper cell types 1 (Th1) and Th17 were reduced, which were validated by in vitro studies. In line with our in vitro data, we found that inflammatory cytokines [tumour necrosis factor‐α (TNF), interleukin ( IL )5, IL 6, IL 1B] were also downregulated in T cells from patients (all P   〈  0·05). Finally, we showed that ruxolitinib does not interfere with the T cell receptor signalling pathway, but impacts IL 2‐dependent STAT 5 activation. These data provide a rationale for testing JAK inhibitors in diseases triggered by hyperactive CD 4 + T cells, such as autoimmune diseases. In addition, they also provide a potential explanation for the increased infection rates (i.e. viral reactivation and urinary tract infection) seen in ruxolitinib‐treated patients.
    Materialart: Online-Ressource
    ISSN: 0007-1048 , 1365-2141
    URL: Issue
    RVK:
    Sprache: Englisch
    Verlag: Wiley
    Publikationsdatum: 2015
    ZDB Id: 1475751-5
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    Online-Ressource
    Online-Ressource
    Informa UK Limited ; 2016
    In:  OncoImmunology Vol. 5, No. 2 ( 2016-02), p. e1071009-
    In: OncoImmunology, Informa UK Limited, Vol. 5, No. 2 ( 2016-02), p. e1071009-
    Materialart: Online-Ressource
    ISSN: 2162-402X
    Sprache: Englisch
    Verlag: Informa UK Limited
    Publikationsdatum: 2016
    ZDB Id: 2645309-5
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    Online-Ressource
    Online-Ressource
    Impact Journals, LLC ; 2015
    In:  Oncoscience Vol. 2, No. 8 ( 2015-08-31), p. 677-678
    In: Oncoscience, Impact Journals, LLC, Vol. 2, No. 8 ( 2015-08-31), p. 677-678
    Materialart: Online-Ressource
    ISSN: 2331-4737
    URL: Issue
    Sprache: Englisch
    Verlag: Impact Journals, LLC
    Publikationsdatum: 2015
    ZDB Id: 2907747-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    In: Journal of Neurology, Springer Science and Business Media LLC, Vol. 266, No. 10 ( 2019-10), p. 2465-2474
    Materialart: Online-Ressource
    ISSN: 0340-5354 , 1432-1459
    RVK:
    Sprache: Englisch
    Verlag: Springer Science and Business Media LLC
    Publikationsdatum: 2019
    ZDB Id: 1421299-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3169-3169
    Kurzfassung: Introduction: Ruxolitinib (INCB018424) is the first JAK inhibitor approved for treatment of myelofibrosis (MF). Ruxolitinib-induced reduction of splenomegaly and symptoms control is linked to a substantial suppression of MF-associated circulating pro-inflammatory and pro-angiogenic cytokines. However, an increased rate of infections in ruxolitinib-exposed patients with MF was recently described. Natural killer (NK) cells are innate immune effector cells eliminating malignant or virus-infected cells. Thus, the aim of this project was to define in more detail the impact of JAK inhibition on NK cell biology both in vitro and in vivo. Methods: 28 patients with myeloproliferative neoplasms (MPN) with or without ruxolitinib therapy and 12 healthy donors were analyzed for NK cell frequency, NK receptor expression and function. Phenotypic and functional NK cell markers (e.g. CD11b, CD27, KIR, NKG2A, NKG2D, NKp46, CD16, granzyme B, and perforin) were analyzed by FACS. NK cell function was evaluated by classical killing assays upon stimulation with MHC class I-deficient target cells K562. Finally, a set of additional in vitro experiments (e.g. analysis of lytic synapse formation by FACS and confocal microscopy) were performed to define in more detail the characteristics and potential mechanisms of ruxolitinib-induced NK cell dysfunction. Results: In addition to our recent finding that ruxolitinib induces NK cell dysfunction in vitro (e.g. reduced killing, degranulation and IFN-γ production), we here demonstrate that NK cell proliferation and cytokine-induced receptor expression as well as cytokine signalling are drastically impaired by ruxolitinib. Interestingly, reduced killing is at least in part due to a reduced capacity to form a mature lytic synapse with target cells. The significance of the in vitrofindings is underscored by a dramatically reduced proportion and absolute number of NK cells in ruxolitinib-treated MPN patients when compared to treatment-naïve patients or to healthy controls (mean percentage of NK cell frequency: ruxolitinib-naïve MPN patients 12.63% ±1.81; healthy donors 13.51% ±1.44; ruxolitinib-treated patients 5.47% ±1.27). A systematic analysis of NK cell receptor expression revealed that the reduction of NK cells in ruxolitinib-exposed individuals is most likely due to an impaired NK cell differentiation and maturation process, as reflected by a significantly increased ratio of immature to mature NK cells. Finally, the endogenous functional NK cell defect in MPN is further aggravated by intake of the JAK inhibitor ruxolitinib. Conclusion: We here provide compelling in vitro and in vivo evidence that inhibition of the JAK/STAT-pathway by ruxolitinib exerts substantial effects on the NK cell compartment in MPN patients due to the inhibition of NK cell differentiation and NK cell key functions. Our data may help to better understand the increased rate of severe infections and complement recent reports on ruxolitinib-induced immune dysfunction. Disclosures Koschmieder: Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Travel, Accomodation, Expenses Other. Brümmendorf:Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties, Research Funding. Wolf:Novartis: Consultancy, Honoraria, Research Funding, Travel and Accommodation Other.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2014
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 8
    Online-Ressource
    Online-Ressource
    American Society of Hematology ; 2015
    In:  Blood Vol. 126, No. 23 ( 2015-12-03), p. 3423-3423
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3423-3423
    Kurzfassung: Introduction: Ruxolitinib is a JAK inhibitor that was recently approved for treatment of primary and secondary MF and shows impressive symptom control by suppression of inflammation. The observed effects are irrespective of the JAK mutational status. Ruxolitinib is also a promising drug for treatment of acute and chronic GvHD (Spoerl et al., Blood 2014 and Zeiser et al., Leukemia, in press). We recently demonstrated that the immune-modulatory effects of ruxolitinib might at least in part be due to an inhibitory effect on dendritic cell (DC) biology (Heine et al., Blood 2014). Dendritic cells are important antigen-presenting cells, which mature and migrate from the periphery into draining lymph nodes upon antigen contact to prime T cells. This process follows chemokine gradients and requires cytoskeletal rearrangement to allow cell movement along the gradients. Members of the family of small GTPases are critically involved in cell chemotaxis, migration and formation of the immunological synapse allowing DC-T cell interaction. The aim of this study was to define in detail the impact of the JAK inhibitor ruxolitinib on DC migration. Methods: CD14+ cells isolated from human buffy coats were differentiated for seven days in the presence of GM-CSF and IL-4 to moDCs and finally matured with LPS. Murine bone marrow-derived DCs (bmDCs) were generated by flushing bone marrow from femura and tibiae of mice and subsequent plating of the cells in medium containing GM-CSF followed by final maturation by LPS. Migration of DCs was analyzed in Transwell assays or dynamically by time-lapse microscopy within three dimensional collagen gels towards CCL19 gradients. Adhesion of cells was tested on different substrates and phenotypic DC marker checked by flow cytometry. Signaling events were analyzed by Western Blot to evaluate changes in phosphorylation levels of relevant proteins of the RhoA/ROCK-pathway. Results: 2D-migration of ruxolitinib-exposed DC on fibronectin is dose-dependently reduced in vitro. By analyzing the migratory phenotype of human moDCs within 3D-collagen gels, ruxolitinib-exposed DCs are still able to sense chemokine gradients as they form lamellipodia at the leading edge of the cell, whereas the retraction of the uropod is clearly inhibited. As a consequence, cell velocity, accumulated and euclidean distance are all significantly reduced by ruxolitinib. These effects are already seen 30 minutes after ruxolitinib exposure. Notably (and in line with the morphological chemokine-sensing by lamellipodia formation), the surface expression of the CCL19-sensing chemokine receptor CCR7 is not altered by ruxolitinib. Moreover, we could not detect any changes of integrin expression or the adhesion of mature DC to fibronectin or collagen as a potential reason for lowered migration due to increased "stickiness" of the DCs. Additional in vivo studies could show that the JAK inhibitor potently reduces homing of bmDCs into draining lymph nodes in mice. Notably, the impaired DC migration is independent of JAK inhibition, as siRNA knockdown experiments revealed that DCs with JAK1, JAK2 or JAK1/2 knockdown migrate appropriately in vitro applying again the 3D-migration system. On a molecular level we could show a reduced phosphorylation of myosin-light chain phosphatase 1 (MYPT1), a direct target of the Rho-associated protein kinase (ROCK) in ruxolitinib-treated moDC upon CCL19 stimulation. Interestingly, the known inhibition of DC activation and maturation seen after treatment with ruxolitinib could not be observed if cells are incubated with the ROCK inhibitor Y-27632. Whereas pharmacological inhibition of ROCK mimicked the migration phenotype seen in moDC exposed to ruxolitinib. Conclusion: RhoA/ROCK pathway is critical for cell migration, as ROCK is a downstream effector of RhoA and leads to stabilization of the actin cytoskeleton via cofilin and acto-myosin II contraction by the myosin light chain. The observed reduction of ROCK activity may reveal an important novel mechanism of ruxolitinib-induced inhibition of DC migration. Our current efforts focus on the validation of ROCK as off-target JAK1/2-independent target kinase of ruxolitinib as potential mediator of the observed DC inhibitory effects, which may at least in part also explain the potential therapeutic impact of ruxolitinib for treatment of GvHD or autoimmunity. Disclosures No relevant conflicts of interest to declare.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2015
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 11 ( 2015-06-01), p. 2187-2199
    Kurzfassung: Ruxolitinib is a small-molecule inhibitor of the JAK kinases, which has been approved for the treatment of myelofibrosis, a rare myeloproliferative neoplasm (MPN), but clinical trials are also being conducted in inflammatory-driven solid tumors. Increased infection rates have been reported in ruxolitinib-treated patients, and natural killer (NK) cells are immune effector cells known to eliminate both virus-infected and malignant cells. On this basis, we sought to compare the effects of JAK inhibition on human NK cells in a cohort of 28 MPN patients with or without ruxolitinib treatment and 24 healthy individuals. NK cell analyses included cell frequency, receptor expression, proliferation, immune synapse formation, and cytokine signaling. We found a reduction in NK cell numbers in ruxolitinib-treated patients that was linked to the appearance of clinically relevant infections. This reduction was likely due to impaired maturation of NK cells, as reflected by an increased ratio in immature to mature NK cells. Notably, the endogenous functional defect of NK cells in MPN was further aggravated by ruxolitinib treatment. In vitro data paralleled these in vivo results, showing a reduction in cytokine-induced NK cell activation. Further, reduced killing activity was associated with an impaired capacity to form lytic synapses with NK target cells. Taken together, our findings offer compelling evidence that ruxolitinib impairs NK cell function in MPN patients, offering an explanation for increased infection rates and possible long-term side effects associated with ruxolitinib treatment. Cancer Res; 75(11); 2187–99. ©2015 AACR.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2015
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 10
    In: Alzheimer's & Dementia: Diagnosis, Assessment & Disease Monitoring, Wiley, Vol. 11, No. 1 ( 2019-12), p. 610-618
    Kurzfassung: Impaired long‐term memory is a defining feature of mild cognitive impairment (MCI). We tested whether this impairment is item specific, limited to some memoranda, whereas some remain consistently memorable. Methods We conducted item‐based analyses of long‐term visual recognition memory. Three hundred ninety‐four participants (healthy controls, subjective cognitive decline [SCD], and MCI) in the multicentric DZNE‐Longitudinal Cognitive Impairment and Dementia Study (DELCODE) were tested with images from a pool of 835 photographs. Results We observed consistent memorability for images in healthy controls, SCD, and MCI, predictable by a neural network trained on another healthy sample. Looking at memorability differences between groups, we identified images that could successfully categorize group membership with higher success and a substantial image reduction than the original image set. Discussion Individuals with SCD and MCI show consistent memorability for specific items, while other items show significant diagnosticity. Certain stimulus features could optimize diagnostic assessment, while others could support memory.
    Materialart: Online-Ressource
    ISSN: 2352-8729 , 2352-8729
    Sprache: Englisch
    Verlag: Wiley
    Publikationsdatum: 2019
    ZDB Id: 2832898-X
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...