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 ; 2013
    In:  Cytokine Vol. 63, No. 3 ( 2013-09), p. 243-
    In: Cytokine, Elsevier BV, Vol. 63, No. 3 ( 2013-09), p. 243-
    Type of Medium: Online Resource
    ISSN: 1043-4666
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2013
    detail.hit.zdb_id: 1463198-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Frontiers Media SA ; 2018
    In:  Frontiers in Endocrinology Vol. 9 ( 2018-6-19)
    In: Frontiers in Endocrinology, Frontiers Media SA, Vol. 9 ( 2018-6-19)
    Type of Medium: Online Resource
    ISSN: 1664-2392
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2018
    detail.hit.zdb_id: 2592084-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal of Extracellular Vesicles, Wiley, Vol. 10, No. 12 ( 2021-10)
    Abstract: Extracellular vesicles (EVs) are characterized by complex cargo composition and carry a wide array of signalling cargo, including growth factors (GFs). Beyond surface‐associated GFs, it is unclear if EV intralumenal growth factors are biologically active. Here, bone morphogenetic protein‐2 (BMP2), loaded directly into the lumen of EVs designated engineered BMP2‐EVs (eBMP2‐EVs), was comprehensively characterized including its regulation of osteoblastogenesis. eBMP2‐EVs and non‐EV ‘free’ BMP2 were observed to similarly regulate osteoblastogenesis. Furthermore, cell trafficking experiments suggest rapid BMP2 recycling and its extracellular release as ‘free’ BMP2 and natural occurring BMP2‐EVs (nBMP2‐EVs), with both being osteogenic. Interestingly, BMP2 occurs on the EV surface of nBMP2‐EVs and is susceptible to proteolysis, inhibition by noggin and complete dissociation from nBMP2‐EVs over 3 days. Whereas, within the eBMP2‐EVs, BMP2 is protected from proteolysis, inhibition by noggin and is retained in EV lumen at 100% for the first 24 h and ∼80% after 10 days. Similar to ‘free’ BMP2, bioprinted eBMP2‐EV microenvironments induced osteogenesis in vitro and in vivo in spatial registration to the printed patterns. Taken together, BMP2 signalling involves dynamic BMP2 cell trafficking in and out of the cell involving EVs, with distinct differences between these nBMP2‐EVs and eBMP2‐EVs attributable to the BMP2 cargo location with EVs. Lastly, eBMP2‐EVs appear to deliver BMP2 directly into the cytoplasm, initiating BMP2 signalling within the cell, bypassing its cell surface receptors.
    Type of Medium: Online Resource
    ISSN: 2001-3078 , 2001-3078
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2021
    detail.hit.zdb_id: 2683797-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 753-753
    Abstract: Multiple myeloma (MM) is a clonal malignancy of plasma cells that frequently causes skeleton destruction. The osteolytic lesions rarely heal even in those patients who are in long-term remission. In MM bone disease, differentiation of osteoblasts is suppressed due to the down-regulation of the Runx2 gene, a master osteoblast differentiation transcription factor. Growth independent factor 1 (Gfi1) is a 55 kDa protein that contains an N-terminal SNAG domain and 6 zinc finger domains at the C-terminus. It is a transcription factor that can recruit chromatin modifiers to genes. We reported that MM cells induce expression of Gfi1 in bone marrow stromal cells, and that Gfi1 directly represses the Runx2 gene. Our previous data indicated that a histone deacetylases (HDAC) inhibitor, Trichostatin A, could block Gfi1-mediated Runx2 promoter repression. However, how Gfi1 activity is modulated by HDAC inhibitors and whether Gfi1 acetylation is involved in the HDAC inhibitor prevention of Runx2 repression are unknown. Therefore, we investigated if Gfi1 is acetylated and if the activity of Gfi1 is regulated by its acetylation status. We also examined the MM-induced changes in Gfi1 and HDACs occupancy on the Runx2 gene. Methods Full length or truncated Gfi1 cDNA constructs were used to express Gfi1 protein by transient transfections. After immunoprecipitation of Gfi1, the acetylation status of Gfi1 was determined by anti-acetylated lysine antibody using western blotting. Overexpression or knockdown of lysine acetyltransferase p300 was done by co-transfection of p300 cDNA or shRNA plasmids. Trichostatin A (5 µM) and nicotinamide (10 mM) were used to inhibit the activity of HDACs. Site-directed mutagenesis was used to change Gfi1 residue lysine 292 to arginine (Gfi1-K292R). For luciferase reporter assays, the pGL4.10 vector constructed with a mouse Runx2 promoter region (-976 ∼ +111) was transfected into mouse pre-osteoblast cell line MC-4. A biotinylated double-stranded oligonucleotide spanning the Gfi1 binding region (-40 ∼ -1) in the mouse Runx2 promoter was utilized for oligonucleotide pull-down assays. Chromatin immunoprecipitation (ChIP) was used to characterize the MM-induced changes in the Runx2 gene chromatin. Results We found that Gfi1 was a target of acetylation and that p300 was co-localized and co-immunoprecipitated with Gfi1. Interestingly, the deacetylation of Gfi1 was controlled by both Zn2+ and NAD+ dependent deacetylases. Using different truncated Gfi1 cDNA constructs, we identified lysine 292 in Gfi1 zinc finger 2 as a critical site for acetylation. Mutant Gfi1-K292R displayed significant resistance to acetylation in the presence of HDAC inhibitors, confirming that Gfi1-K292 is the major acetylation site. Importantly, oligonucleotide pull down assays showed that Gfi1-K292R exhibited stronger DNA binding activity than Gfi1-WT to the Gfi1 binding site in the Runx2 promoter. In luciferase assays, increasing or decreasing lysine acetyltransferase activity by overexpression or knockdown of p300 resulted in the attenuation or enhancement in Gfi1 repression of the Runx2 promoter, respectively. Further, the acetylation-resistant Gfi1-K292R more strongly repressed the Runx2 promoter, which was not rescued by HDAC inhibitors, indicating that the contribution of HDAC inhibitors to increased acetylated Gfi1 is a major mechanism for prevention of Gfi1-mediated Runx2 repression. Lastly, ChIP analysis of the Runx2 gene in MC-4 cells before and after co-culture with myeloma cells revealed that increased Gfi1 occupancy at the Runx2 gene correlated with increased HDAC1 and decreased Pol-II and H3K9ac, confirming that the interaction between Gfi1 and HDACs is involved in the repression of the Runx2 gene. Conclusions Taken together, our results indicate that Gfi1 is an acetylated protein and suggest that increasing acetylation of Gfi1represents a potential therapeutic target for reversing Gfi1-mediated Runx2 repression and promoting osteoblast differentiation in MM bone disease. Disclosures: Roodman: Amgen: Membership on an entity’s Board of Directors or advisory committees; Eli Lilly: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 3247-3247
    Abstract: Multiple myeloma (MM) is the most frequent cancer to involve the skeleton with patients developing osteolytic bone lesions due to hyperactivation of osteoclasts (OCL) resulting in severe bone pain, pathological fractures and enhanced mortality. These bone lesions rarely heal even after therapeutic remission due to MM-induced suppression of bone marrow stromal cells (BMSC) differentiation into functional osteoblasts (OB), enhancing their support of MM growth and survival. Although new therapies for MM have greatly improved progression-free survival and overall survival, MM remains incurable for most patients. EZH2is the methyltransferase subunit of the Polycomb Repressive Complex 2 (PRC2) catalyzing the tri-methylation of histone-3 lysine-27 (H3K27me3), which induces gene repression. We previously reported that MM cells induce increased recruitment of EZH2 to the Runx2 gene in the murine pre-OB cell line MC4 resulting in H3K27me3-mediated repression of Runx2, thereby causing repression of OB differentiation. In the present study, we show that the repressive H3K27me3 levels are elevated with reduced H3K9 acetylation on the Runx2 promoter in MM patient derived BMSC. Using the selective small molecule inhibitor GSK126 to block EZH2 activity, we investigated if the MM-induced epigenetic repression of Runx2 is reversible. We found that GSK126 enhances the active architecture at the Runx2 promoter in both BMSC from MM patients and MM-treated MC4 cells, and rescues the osteogenic potential and mineralization capability. 5TGM1 MM cells exhibited reduced adhesion to MC4 pre-OB pre-treated with GSK126 as compared to vehicle in an in vitro adhesion assay, suggesting that EZH2 inhibition will decrease pre-OB support for MM cells. In addition to the direct osteo-anabolic effects of EZH2 inhibition on OB differentiation, we investigated if GSK126 treatment affected the pro-inflammatory, myeloma-amplified OCL differentiation that results in enhanced osteolysis in MM. We reported that expansion of bone marrow monocytes (BMM), which are OCL precursors, in the presence of MM1.S-conditioned media, significantly enhanced formation of TRAP-positive multinucleated OCL. Here we show that MM-conditioned media significantly increased EZH2 and OCL marker genes NFATc1, RANK, OSCAR, Cathepsin K, and DC-STAMP mRNA and enhanced RANKL-induced formation of OCL, which was blocked by GSK126. We found that EZH2 and the corresponding H3K27me3 levels increase during the initial 24h of RANKL-induced osteoclastogenesis and that GSK126 inhibition during the first 24h of RANKL treatment was necessary and sufficient to decrease formation of mature OCL. Thus, GSK126 is likely to both protect the bone from new lytic lesions and help heal the existing osteolytic lesions in MM. We found that GSK126 treatment (48h) inhibited proliferation of human (MM1.S, RPMI, U266, H929, and JJN3) and murine (5TGM1) MM cell lines, and primary 138+MM patient cells. We analyzed the interaction of GSK126 and bortezomib using IC50 constant ratio drug combinations based on the Chou & Talalay method using the CompuSyn program. We found that GSK126 synergizes with bortezomib to inhibit cell viability of these MM cell lines and activated apoptosis via induction of activated caspase-3. We used 5TGM1-GLuc-GFP cells, which secrete Guassia luciferase (GLuc), to measure MM cell viability in a longitudinal drug treatment study. GLuc levels in media correlated with cell proliferation, which was inhibited by single and combination doses of GSK126 and bortezomib, further demonstrating the synergism of these drugs. GSK126 decreased EZH2 mRNA and protein levels in MM cells, which is a cell-type specific effect, since we did not observe EZH2 protein alteration in GSK126 treated OCL progenitors. Our in vitro study suggests that selective inhibition of the epigenetic modifier EZH2 using GSK126 would positively regulate the bone microenvironment by improving the osteogenic potential of MM-exposed BMSC and suppressing OCL formation, and would directly decrease MM cell survival, particularly in combination with bortezomib. These data suggest that targeting EZH2 activity alone or in combination with bortezomib may prove a valuable therapeutic strategy to improve bone health and limit disease progression in MM patients. Disclosures Roodman: Amgen: Consultancy.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 4216-4216
    Abstract: Multiple myeloma (MM) causes osteolytic bone lesions that rarely heal even after therapeutic remission. We reported that the MM-induced pro- inflammatory bone marrow microenvironment causes upregulation of the transcriptional repressor Gfi1 in bone marrow stromal cells (BMSC) via TNFα. Gfi1 represses the key osteoblast (OB) differentiation factor Runx2, resulting in impaired BMSC differentiation into OB. In this study, we explored the molecular mechanisms involved in Gfi1-mediated repression of Runx2 and the role of histone modifiers and possibly of cofactors, which co-operate with the MM-induced Gfi1 binding to epigenetically repress Runx2. The Runx2 promoter in MC4 pre-OB co-cultured with 5TGM1 MM cells (48 h) had reduced activating acetylation (H3K9ac) levels and enhanced heterochromatic methylation (H3K27me3), consistent with decreased mRNA expression that stayed refractory to OB differentiation. BMSC from MM patients compared to normals also had decreased levels of H3K9Ac at the Runx2 gene promoter. MM co-culture induced binding of Gfi1 to the Runx2 promoter in pre-OB with increased occupancy at 36 and 48 h, concomitantly with increased histone deacetylase HDAC1 (erases H3K9ac) and the PRC2 complex methyltransferase subunit EZH2 (adds H3K27me3). Further, ectopic Gfi1 also bound Runx2 and recruited these histone modifiers. We found that Gfi1 knockdown in pre-OB MC4 prevented MM-dependent HDAC1 and EZH2 recruitment, resulting in less Runx2 de-acetylation, lack of repressive H3K27 tri-methylation and rescue of differentiation-induced Runx2 mRNA expression. Additionally, the use of the selective inhibitors MC1293 (HDAC1i) and GSK126 (EZH2i) also prevented Runx2 mRNA suppression in MM treated pre-OB. The LIM-domain protein family member Ajuba was reported to serve as a Gfi1 corepressor on a subset of Gfi1 target genes in macrophages. Therefore, we investigated if Gfi1 requires Ajuba to repress Runx2 in pre-OB. After 36-48 h MM exposure of MC4 cells, enrichment of Ajuba occupancy was co-localized to the Gfi1 binding site in the Runx2 promoter concurrently with the recruitment of Gfi1. Biotin-oligo pulldown of Gfi1 brought down Ajuba as well. Further, co-transfection of Ajuba and Gfi1 revealed that Ajuba enhanced repression by suboptimal doses of Gfi1 of both a Runx2-luciferase reporter as well as the endogenous Runx2 gene in pre-OB. Studies using deletion constructs showed that the LIM region of Ajuba in conjunction with Gfi1 is necessary and sufficient for Runx2 repression, and the pre-LIM portion of Ajuba does not affect Runx2 luciferase expression. Transfected Ajuba exhibits cytoplasmic localization in MC4 cells unless co-expressed with full-length Gfi1, which brings it into the nucleus. Nuclear co-localization of Ajuba with Gfi1 was uncoupled in MC4 cells when Ajuba was co-transfected with Gfi1 containing only the DNA binding region (aa 239-423). Transfected 239-423 Gfi1 binds the endogenous Runx2 promoter, but fails to repress transcription, likely due to impaired recruitment of Ajuba and histone co-repressors. Importantly, knockdown of Ajuba caused decreased recruitment of Gfi1 to the Runx2 gene in pre-OB and prevented the Gfi1 repression of a Runx2 reporter. Collectively these data show that Ajuba functions as a required Gfi1 co-factor recruiting HDAC1 and EZH2 to establish long-termepigenetic suppression of Runx2 transcription in OB lineage cells in MM bone disease. Disclosures Roodman: Amgen: Consultancy; Eli Lilly: Research Funding.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Wiley ; 2010
    In:  The FASEB Journal Vol. 24, No. S1 ( 2010-04)
    In: The FASEB Journal, Wiley, Vol. 24, No. S1 ( 2010-04)
    Type of Medium: Online Resource
    ISSN: 0892-6638 , 1530-6860
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2010
    detail.hit.zdb_id: 1468876-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 12 ( 2020-12-01), p. 1554-1567
    Abstract: Therapeutic cancer vaccines targeting melanoma-associated antigens are commonly immunogenic but are rarely effective in promoting objective clinical responses. To identify critical molecules for activation of effective antitumor immunity, we have profiled autologous dendritic cell (DC) vaccines used to treat 35 patients with melanoma. We showed that checkpoint molecules induced by ex vivo maturation correlated with in vivo DC vaccine activity. Melanoma patient DCs had reduced expression of cell surface inducible T-cell costimulator ligand (ICOSL) and had defective intrinsic NF-κB signaling. Chromatin immunoprecipitation assays revealed NF-κB–dependent transcriptional regulation of ICOSL expression by DCs. Blockade of ICOSL on DCs reduced priming of antigen-specific CD8+ and CD4+ T cells from naïve donors in vitro. Concentration of extracellular/soluble ICOSL released from vaccine DCs positively correlated with patient clinical outcomes, which we showed to be partially regulated by ADAM10/17 sheddase activity. These data point to the critical role of canonical NF-κB signaling, the regulation of matrix metalloproteinases, and DC-derived ICOSL in the specific priming of cognate T-cell responses in the cancer setting. This study supports the implementation of targeted strategies to augment these pathways for improved immunotherapeutic outcomes in patients with cancer.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Bone Abstracts, Bioscientifica, ( 2016-04-21)
    Type of Medium: Online Resource
    ISSN: 2052-1219
    Language: Unknown
    Publisher: Bioscientifica
    Publication Date: 2016
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    Elsevier BV ; 2022
    In:  Trends in Immunology Vol. 43, No. 6 ( 2022-06), p. 438-448
    In: Trends in Immunology, Elsevier BV, Vol. 43, No. 6 ( 2022-06), p. 438-448
    Type of Medium: Online Resource
    ISSN: 1471-4906
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    detail.hit.zdb_id: 2040190-5
    SSG: 12
    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...