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
Filter
  • Online Resource  (862)
  • Li, Xiaoming  (862)
Material
  • Online Resource  (862)
Language
  • 1
    In: EMBO reports, EMBO, Vol. 24, No. 12 ( 2023-12-06)
    Abstract: image T‐cell specific Rip1 KO mice show signs of accelerated aging‐like phenotypes, along with premature death. Blocking apoptosis by co‐deleting Fadd in Rip1 tKO mice significantly rescues aging‐like phenotypes and prolongs shortened lifespan. Rip1 tKO mice show similar age‐related T cell changes and exhibit signs of accelerated aging and a shorter lifespan. Rip1 ‐deficient T cells undergo excessive apoptosis, promoting chronic inflammation. Blocking necroptosis has little effect on the multimorbidity and premature aging seen in Rip1 tKO mice. Blocking T cell apoptosis by co‐deleting Fadd rescues premature aging and multimorbidity in Rip1 tKO mice.
    Type of Medium: Online Resource
    ISSN: 1469-221X , 1469-3178
    Language: English
    Publisher: EMBO
    Publication Date: 2023
    detail.hit.zdb_id: 2025376-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cell Death & Differentiation, Springer Science and Business Media LLC, Vol. 29, No. 10 ( 2022-10), p. 2034-2045
    Type of Medium: Online Resource
    ISSN: 1350-9047 , 1476-5403
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 1496681-5
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal of Clinical Investigation, American Society for Clinical Investigation, Vol. 123, No. 1 ( 2013-1-2), p. 315-328
    Type of Medium: Online Resource
    ISSN: 0021-9738
    Language: English
    Publisher: American Society for Clinical Investigation
    Publication Date: 2013
    detail.hit.zdb_id: 2018375-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 61-61
    Abstract: Abstract 61 To identify new therapeutic strategies for AML, we compiled and screened an in-house library of on-patent and off-patent drugs to identify agents cytotoxic to leukemia cells. From this screen, we identified mefloquine, an off-patent drug indicated for the treatment and prophylaxis of malaria. In secondary assays, mefloquine decreased the viability of 9/10 human and murine leukemia cell lines (EC50 3.25–8.0 μM). Moreover, it reduced the viability of 4/5 primary AML samples, but was not cytotoxic to normal hematopoietic cells (EC50 〉 31 μM). Importantly, mefloquine reduced the clonogenic growth of primary AML samples, but not normal hematopoietic cells, and completely inhibited engraftment of primary AML cells into immune deficient mice. Finally, systemic treatment with oral mefloquine (50 mg/kg/day) decreased leukemic burden without evidence of toxicity in 4 mouse models of leukemia, including mice engrafted with primary AML cells. Thus, mefloquine effectively targets leukemic cells, including leukemia stem cells, at concentrations that appear pharmacologically achievable and are not toxic to normal hematopoietic cells. To identify the mechanisms of mefloquine-mediated cell death in AML cells, we performed a binary drug combination screen, hypothesizing that drugs that synergized with mefloquine may share overlapping mechanism of action. From this combination screen of 550 drugs, we identified 18 that reproducibly synergized with mefloquine as measured by the Excess over Bliss additivism score, including 3 members of the artemisinin class of anti-malarials: artemisinin, artesunate and artenimol. Strikingly, 10/18 synergistic compounds, including the artemisinins, were known generators of reactive oxygen species (ROS). Therefore we tested mefloquine's ability to increase ROS in leukemic cells. Mefloquine increased ROS production in leukemia cells in a dose- and time-dependent manner. Co-treatment with ROS scavengers α-tocopherol and N-acetyl-cysteine abrogated mefloquine-induced ROS production and cell death, indicating that ROS production was functionally important for mefloquine-mediated cell death. Moreover, the artemisinins induced ROS as single agents, and synergistically increased ROS when combined with mefloquine. To identify cellular target(s) of mefloquine's anti-leukemic effects, we performed a yeast genome-wide functional screen to identify heterozygous gene deletions that rendered yeast more sensitive to mefloquine. 21/37 genes whose depletion conferred 〉 4-fold sensitivity to mefloquine were associated with function of the yeast vacuole, equivalent to the mammalian lysosome. Consistent with these data, fluorescent confocal microscopy demonstrated that mefloquine and artesunate disrupted lysosomes. Cell death after mefloquine and artesunate treatment was caspase-independent and associated with increased incorporation of monodancylcadaverin in autophagosomes, consistent with the effect of these drugs on the lysosomes. To further explore the anti-leukemic activity of lysosomal disruption, we evaluated the anti-leukemic effects of the known lysosomal disrupter L-leucine-leucine methyl ether (LeuLeuOMe). Similar to mefloquine and artesunate, LeuLeuOMe induced cell death in leukemia cells, increased ROS production, and disrupted the lysosomes. Highlighting the potential clinical utility of lysosomal disrupters for the treatment of leukemia, a patient with relapsed/refractory juvenile myelomonocytic leukemia self-administered artemisinin. The artemisinin cleared the circulating blasts from the circulating blasts and the patient proceeded to allotransplant. Finally, to investigate the basis of leukemic cell hypersensitivity to lysosomal disruption, we assessed lysosomal characteristics of primary AML and normal hematopoietic cells. By gene expression analysis, AML patient samples had higher mRNA levels of the lysosomal cathepsins A, B, C, D, H, L, S and Z, compared to CD34+ normal hematopoietic cells, and cathepsins C, D and Z were significantly over-expressed in the LSC compartment, compared to normal HSCs. In summary, our data demonstrate that lysosomal disruption preferentially targets AML cells and AML stem cells through a mechanism related to increased ROS production. Thus, this work highlights lysosomal disruption as a novel therapeutic strategy for AML. Disclosures: Off Label Use: This study includes a case report of off-label use of the anti-malarial artemisinin in the treatment of a case of juvenile myelomonocytic leukemia.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    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
    Springer Science and Business Media LLC ; 2015
    In:  Environmental Monitoring and Assessment Vol. 187, No. 4 ( 2015-4)
    In: Environmental Monitoring and Assessment, Springer Science and Business Media LLC, Vol. 187, No. 4 ( 2015-4)
    Type of Medium: Online Resource
    ISSN: 0167-6369 , 1573-2959
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2015
    detail.hit.zdb_id: 2012242-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 1711-1711
    Abstract: The proteasome is an enzymatic complex that rids cells of excess and misfolded proteins and possesses chymotrypin, trypsin, and caspase-like enzymatic activity. To date, all of the proteasome inhibitors approved for clinical use or in clinical trials inhibit the complex competitively by binding the active sites of the enzymes. Here, we report a novel chemical proteasome inhibitor that binds the alpha subunits of the 20S proteasome and inhibits the complex non-competitively through a dual copper-dependent and independent mechanism. In a screen of a focused chemical library for novel proteasome inhibitors, we identified 5-amino-8-hydroxyquinoline (5AHQ). When added to myeloma or leukemia intact cells or cell extracts, 5AHQ inhibited the enzymatic activity of the proteasome at low micromolar concentrations. In order to obtain further insight into the mechanism of action of 5AHQ, we carried out a kinetic analysis of inhibition of the enzymatic activity of purified T. Acidophilium proteasome. By Lineweaver-Burk plot analysis, 5AHQ inhibited the proteasome non-competitively. Next, we investigated the binding of 5AHQ to the proteasome. By NMR analysis, 5AHQ bound the half-proteasome complex comprised of a pair of α-rings, α7-α7, and clear spectral changes were observed that localized to residues Ile159, Val113, Val87, Val82, Leu112, Val89, Val134, Val24 and Leu136 inside the antechamber. In contrast, the competitive inhibitor MG132 that binds the proteolytic chamber did not produce any changes in spectra of α7-α7, as expected. 5AHQ bound copper in a 2:1 stoichiometry with a logβ′ value of 9.09, and the addition of copper to 5AHQ enhanced 5AHQ-mediated inhibition of the proteasome. However, binding intracellular copper was not sufficient to explain the effects of 5AHQ on the proteasome as analogues of 5AHQ that did not bind copper continued to inhibit the proteasome, copper-binding molecules not structurally related to 5AHQ did not affect the proteasome, and 5AHQ inhibited isolated proteasomes in buffers devoid of copper and other heavy metals. Given the effects of 5AHQ on the proteasome, we examined the effects of this molecule on the viability of leukemia and myeloma cell lines. Leukemia, myeloma and solid tumor cell lines were treated with increasing concentrations of 5AHQ for 72 hours and cell viability was measured by the MTS assay. 5AHQ induced cell death in 9/9 myeloma, 6/10 leukemia, and 3/10 solid tumor cell lines with an LD50 ≤5 uM. Cell death was confirmed by Annexin V staining. Consistent with its mechanism of action as a proteasome inhibitor, the ability of 5AHQ to induce cell death matched its ability to inhibit the proteasome. In addition, 5AHQ-mediated cell death was associated with inhibition of the NF-kappaB signalling pathway. As 5AHQ induced cell death in malignant cells, we evaluated the effects of oral 5AHQ in 3 mouse models of leukemia. Sublethally irradiated NOD-SCID mice were injected subcutaneously with OCI-AML2 or K562 human leukemia cells or intraperitoneally with MDAY-D2 murine leukemia cells. After tumor implantation, mice were treated with 5AHQ (50 mg/kg/day) or buffer control by oral gavage. Oral 5AHQ decreased tumor weight and volume in all 3 mouse models compared to control without causing weight loss or gross organ toxicity. In summary, we have identified a new strategy for inhibition of the proteasome and a lead for a new therapeutic agent for the treatment of hematologic malignancies.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    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. 115, No. 11 ( 2010-03-18), p. 2251-2259
    Abstract: The proteasomal pathway of protein degradation involves 2 discrete steps: ubiquitination and degradation. Here, we evaluated the effects of inhibiting the ubiquitination pathway at the level of the ubiquitin-activating enzyme UBA1 (E1). By immunoblotting, leukemia cell lines and primary patient samples had increased protein ubiquitination. Therefore, we examined the effects of genetic and chemical inhibition of the E1 enzyme. Knockdown of E1 decreased the abundance of ubiquitinated proteins in leukemia and myeloma cells and induced cell death. To further investigate effects of E1 inhibition in malignancy, we discovered a novel small molecule inhibitor, 3,5-dioxopyrazolidine compound, 1-(3-chloro-4-fluorophenyl)-4-[(5-nitro-2-furyl)methylene] -3,5-pyrazolidinedione (PYZD-4409). PYZD-4409 induced cell death in malignant cells and preferentially inhibited the clonogenic growth of primary acute myeloid leukemia cells compared with normal hematopoietic cells. Mechanistically, genetic or chemical inhibition of E1 increased expression of E1 stress markers. Moreover, BI-1 overexpression blocked cell death after E1 inhibition, suggesting ER stress is functionally important for cell death after E1 inhibition. Finally, in a mouse model of leukemia, intraperitoneal administration of PYZD-4409 decreased tumor weight and volume compared with control without untoward toxicity. Thus, our work highlights the E1 enzyme as a novel target for the treatment of hematologic malignancies.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    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: ImmunoHorizons, The American Association of Immunologists, Vol. 6, No. 7 ( 2022-07-01), p. 465-475
    Abstract: Caspase-8 (Casp8) suppresses receptor-interacting protein kinase-3 (RIPK3)/mixed lineage kinase domain-like protein (MLKL)-dependent necroptosis, demonstrated by the genetic evidence that deletion of Ripk3 or Mlkl prevented embryonic lethality of Casp8-deficient mice. However, the detailed mechanisms by which Casp8 deficiency triggers necroptosis during embryonic development remain unclear. In this article, we show that Casp8 deletion caused formation of the RIPK1-RIPK3 necrosome in the yolk sac, leading to vascularization defects, prevented by MLKL and RIPK3 deficiency, or RIPK3 RHIM mutant (RIPK3 V448P), but not by the RIPK1 kinase-dead mutant (RIPK1 K45A). In addition, Ripk1K45A/K45ACasp8−/− mice died on embryonic day 14.5, which was delayed to embryonic day 17.5 by ablation of one allele in Ripk1 and was completely rescued by ablation of Mlkl. Our results revealed an in vivo role of RIPK3 RHIM and RIPK1K45A scaffold-mediated necroptosis in Casp8 deficiency embryonic development and suggested that the Casp8-deficient yolk sac might be implicated in identifying novel regulators as an in vivo necroptotic model.
    Type of Medium: Online Resource
    ISSN: 2573-7732
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2022
    detail.hit.zdb_id: 2882729-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: JNCI: Journal of the National Cancer Institute, Oxford University Press (OUP), Vol. 102, No. 14 ( 2010-7), p. 1069-1082
    Type of Medium: Online Resource
    ISSN: 0027-8874 , 1460-2105
    RVK:
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2010
    detail.hit.zdb_id: 2992-0
    detail.hit.zdb_id: 1465951-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 2529-2529
    Abstract: Off patent drugs with previously unrecognized anti-leukemia activity can be rapidly repurposed for this new indication, given their prior safety and toxicity testing. By screening a group of anti-malarial compounds for anti-cancer activity, we identified mefloquine, a quinoline licensed in oral formulation for the treatment of malaria. As an anti-cancer agent, we demonstrated that mefloquine decreased the viability of 9/9 leukemia cell lines with an LD50 & lt;7.5 uM, and 9/9 myeloma cell lines with an LD50 & lt;5.0 uM. Furthermore, mefloquine demonstrated induced cell death in primary AML samples (n = 3; LD50 & lt;7.5 uM), but not normal peripheral blood stem cells. Given its in vitro activity, we evaluated the effects of oral mefloquine in mouse xenograft models of leukemia. Sublethally irradiated SCID mice were injected subcutaneously with OCI-AML2 or K562 human leukemia cells or MDAY-D2 murine leukemia cells, and treated with 50 mg/kg mefloquine, or vehicle alone by oral gavage. Oral mefloquine decreased tumor weight and volume in all 3 mouse models without toxicity. Mechanistically, mefloquine induced reactive oxygen species (ROS) in leukemia cells at times preceding and concentrations associated with cell death. Blockade of ROS by N-acetyl-L-cysteine (NAC) abrogated mefloquine sensitivity, suggesting that mefloquine-mediated cell death in AML was ROS-dependent. To further understand the mechanism of mefloquine-mediated cytoxicity, whole genome gene expression oligonucleotide array analysis of AML cells treated with mefloquine was conducted. The gene expression pattern of cells treated with mefloquine strongly resembled gene signatures associated with activated Toll-like receptor and interferon response pathways. STAT1 and NF-κB, both downstream transcription factor components of TLR-IFN signaling, were activated, as were downstream targets IRF1, IRF7 and IL-8, at times that preceded mefloquine-induced cell death. Gene expression changes were validated by Q-RT-PCR, and are potential biomarkers of mefloquine activity in cells. This pathway appears functionally important for mefloquine-mediated cell death, as cell lines defective for STAT1 signaling components showed decreased cell death after mefloquine treatment. These lines also showed decreased ability to generate ROS in response to mefloquine treatment, suggesting that mefloquine-induced ROS was produced through a STAT1-dependent mechanism. Taken together, our data demonstrate that the anti-malarial mefloquine displays significant pre-clinical activity in leukemia and myeloma cells, likely through a STAT1-dependent induction of ROS that is triggered by activation of the TLR-IFN cytokine axis in cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2529.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    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...