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: Parasitology, Cambridge University Press (CUP), Vol. 150, No. 8 ( 2023-07), p. 661-671
    Abstract: Trematodes of the genus Ogmocotyle are intestinal flukes that can infect a variety of definitive hosts, resulting in significant economic losses worldwide. However, there are few studies on molecular data of these trematodes. In this study, the mitochondrial (mt) genome of Ogmocotyle ailuri isolated from red panda ( Ailurus fulgens ) was determined and compared with those from Pronocephalata to investigate the mt genome content, genetic distance, gene rearrangements and phylogeny. The complete mt genome of O. ailuri is a typical closed circular molecule of 14 642 base pairs, comprising 12 protein-coding genes (PCGs), 22 transfer RNA genes, 2 ribosomal RNA genes and 2 non-coding regions. All genes are transcribed in the same direction. In addition, 23 intergenic spacers and 2 locations with gene overlaps were determined. Sequence identities and sliding window analysis indicated that cox 1 is the most conserved gene among 12 PCGs in O. ailuri mt genome. The sequenced mt genomes of the 48 Plagiorchiida trematodes showed 5 types of gene arrangement based on all mt genome genes, with the gene arrangement of O. ailuri being type I. Phylogenetic analysis using concatenated amino acid sequences of 12 PCGs revealed that O. ailuri was closer to Ogmocotyle sikae than to Notocotylus intestinalis . These data enhance the Ogmocotyle mt genome database and provide molecular resources for further studies of Pronocephalata taxonomy, population genetics and systematics.
    Type of Medium: Online Resource
    ISSN: 0031-1820 , 1469-8161
    RVK:
    Language: English
    Publisher: Cambridge University Press (CUP)
    Publication Date: 2023
    detail.hit.zdb_id: 1491287-9
    detail.hit.zdb_id: 207627-5
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    The Society of Laparoscopic and Robotic Surgeons ; 2013
    In:  JSLS : Journal of the Society of Laparoendoscopic Surgeons Vol. 17, No. 2 ( 2013), p. 263-272
    In: JSLS : Journal of the Society of Laparoendoscopic Surgeons, The Society of Laparoscopic and Robotic Surgeons, Vol. 17, No. 2 ( 2013), p. 263-272
    Type of Medium: Online Resource
    ISSN: 1086-8089 , 1938-3797
    Language: Unknown
    Publisher: The Society of Laparoscopic and Robotic Surgeons
    Publication Date: 2013
    detail.hit.zdb_id: 2011211-7
    detail.hit.zdb_id: 2238601-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 2890-2890
    Abstract: Leukemic Stem Cells (LSCs) isolated from Acute Myeloid Leukemia (AML) patients are highly sensitive to NF-κB inhibition-induced cell death in in vitro culture when compared to normal hematopoietic stem/progenitor cells (HSPCs). This suggests blocking NF-κB could be an effective strategy for treating AML. However, NF-κB inhibitor treatment alone is unable to clear AML tumors in vivo. We propose this is most likely due to elements within the niche microenvironment stimulating compensatory survival/proliferation signals in AML cells that can overcome NF-κB inhibition. We utilize a multi-faceted AML model that includes a murine system created by MLL-AF9 transduced hematopoietic cells, several established human AML cell lines, and primary AML tumors isolated from patients. We observed elevated Tumor Necrosis Factor-α (TNF) levels in the peripheral blood of AML M3, 4, and 5 patients. Our studies suggest this TNF is produced directly by the AML cells, and that AML cells utilize TNF to stimulate JNK as a survival signal parallel to NF-κB in leukemic cells and a death signal in HSPCs. In healthy HSPCs, stimulation with exogenous TNF induces RIP1/3-mediated necroptosis and caspase-mediated apoptosis, resulting in cell death and loss of normal hematopoietic function. Inactivation of JNK signaling can partially rescue TNF-mediated loss of function in HSPCs. Additionally, blocking NF-κB in HSPCs sensitizes them to TNF-mediated cell death, which can also be partially prevented by inactivation of JNK signaling. These data suggest that JNK acts as a TNF-mediated death signal in HSPCs. In AML, we found TNF signaling promotes the in vitro growth of leukemic cells as well as enhancing the in vivo development of AML tumors by stimulation of both NF-κB and JNK signaling in parallel. We determined that TNF-mediated JNK signaling in leukemic cells results in the propagation of a critical survival signal. These cells convert TNF-JNK stimulation from a death to survival signal by limiting JNK activity duration as well as upregulating the expression of the AP1 family transcription factor c-Jun. Therefore, TNF-JNK signaling in leukemic cells does not drive apoptotic cell death, but instead drives c-Jun activity and the subsequent production of the anti-apoptotic genes c-Flip and Mcl-1. We found inactivation of any part of the TNF-JNK-AP1 signaling axis can repress the growth of leukemic cells in vitro and delay leukemogenesis in vivo. We also show that blocking any portion of the TNF-JNK-AP1 signaling axis sensitizes TNF-expressing leukemic cells, including LSCs, to NF-κB inhibitor treatment while at the same time protecting HSPCs from such treatment. In conclusion, our studies suggest that the inadequate ability of NF-κB inhibition to clear AML tumors in vivo is due to the TNF-mediated activation of the JNK-AP1 signaling axis that can compensate for NF-κB signal repression in LSCs. We found that many types of AMLs produce TNF, and this TNF acts both in an autocrine fashion to promote LSC survival and self-renewal through parallel activation of NF-κB and JNK-AP1, as well as in a paracrine fashion to repress normal hematopoiesis. Therefore, we propose that inhibition of both TNF-JNK-AP1 and NF-κB signals may provide a more thorough treatment for AML patients with elevated peripheral blood TNF. 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: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 118, No. 23 ( 2011-12-01), p. 6057-6067
    Abstract: We studied the effects of TNF-α and Fas-induced death signaling in hematopoietic stem and progenitor cells (HSPCs) by examining their contributions to the development of bone marrow failure syndromes in Tak1-knockout mice (Tak1−/−). We found that complete inactivation of TNF-α signaling by deleting both of its receptors, 1 and 2 (Tnfr1−/−r2−/−), can prevent the death of 30% to 40% of Tak1−/− HSPCs and partially repress the bone marrow failure phenotype of Tak1−/− mice. Fas deletion can prevent the death of 5% to 10% of Tak1−/− HSPCs but fails to further improve the survival of Tak1−/−Tnfr1−/−r2−/− HSPCs, suggesting that Fas might induce death within a subset of TNF-α-sensitive HSPCs. This TNF-α/Fas-induced cell death is a type of receptor-interacting protein-1 (RIP-1)–dependent programmed necrosis called necroptosis, which can be prevented by necrostatin-1, a specific RIP-1 inhibitor. In addition, we found that the remaining Tak1−/− HSPCs died of apoptosis mediated by the caspase-8–dependent extrinsic apoptotic pathway. This apoptosis can be converted into necroptosis by the inhibition of caspase-8 and prevented by inhibiting both caspase-8 and RIP-1 activities. We concluded that HSPCs are heterogeneous populations in response to death signaling stimulation. Tak1 mediates a critical survival signal, which protects against both TNF-α/Fas-RIP-1–dependent necroptosis and TNF-α/Fas-independent apoptosis in HSPCs.
    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
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 3714-3714
    Abstract: Background: Among all tissues, bone marrow (BM) is the most sensitive tissue to ionizing radiation (IR)-induced acute tissue damage (ATD) and chronic long-term residual damage (LT-RD). BM failure and a significant reduction in blood cells (pancytopenia) often occurs within days after exposure to IR due to the massive death of proliferative hematopoietic progenitor cells (HPCs). However, due to their quiescent cell cycle status and reduced fidelity of DNA repair feature, many hematopoietic stem cells (HSCs) cannot fully eliminate such damage and enter senescence; this results in LT-RD. Abnormal dysplastic hematopoiesis is the most common LT-RD in most victims of IR, followed by an increased risk of leukemia/lymphoma development. Thus IR exposure is an established cause of BM failure and leukemia. A significant increase in the production of inflammatory cytokines is induced by IR which contributes to the pathogenesis of both ATD and LT-RD. Such inflammatory cytokines induce the activation of Ripk3-Mlkl-mediated necroptotic signaling in HSCs. However, the role of Ripk3-Mlkl signaling in IR-induced damage has not studied. Experimental procedures: The self-renewal capacity of HSCs among Ripk3-/-, Mlkl-/- and WT mice were examined and compared by serial transplantation assay. The phenotypes of ATD and LT-RD induced by different dosages of IR were compared among Ripk3-/-, Mlkl-/- and WT mice. The mechanism by which Ripk3 signaling prevents IR-induced leukemia development was studied. Results: Ripk3-Mlkl signaling is not required for hematopoiesis during homeostatic condition. However, during serial transplantation, inactivation of such signaling prevents stress-induced loss of HSCs. Interestingly, Ripk3 signaling also induces an Mlkl-independent ROS-p38-p16-mediated senescence in HSCs. Thus Ripk3-/- HSCs showed better competitive hematopoietic ability compared to Mlkl-/- and WT HSCs during serial transplantation. A sub-lethal dosage of IR (6Gy) induces Ripk3-dependent NF-κB activation and pro-survival gene expression in HSCs, which is necessary for the survival of damaged HSCs. After 6Gy IR, although DNA damage is repaired in most HSCs within 2 days, a proportion of HSCs in WT and Mlkl-/- mice fail to fully repair the damage and undergo p53-p21-dependent senescence. However such cells in Ripk3-/- mice die from apoptosis. Thus the remaining HSCs in Ripk3-/- mice should be functionally normal, while a proportion of the remaining HSCs in Mlkl-/- and WT mice remain damaged but senescent, all as demonstrated by competitive hematopoietic reconstitution assay. Multiple low-doses of IR (1.75Gy once week × 4) induce HSC exhaustion in WT mice but not in Ripk3-/- and Mlkl-/- mice. Interestingly, almost all Ripk3-/- mice develop acute lymphoblastic leukemia within 200 days after such low dose IR, while 45% of WT and 60% of Mlkl-/- mice develop thymomas within 360 days (see Figure). Mechanistically, such low-dose IR stimulates chronic inflammatory cytokine production. Such cytokines induce Ripk3-Mlkl-mediated necroptosis in response to HSC exhaustion observed in WT mice. These cytokines also induce Ripk3-ROS-p38-p16-mediated senescence in response to impaired HSC functioning observed in both WT and Mlkl-/- mice. In Ripk3-/- mice, due to the lack of both necroptotic and senescent signaling, mutant HSCs accumulate and leukemia development is accelerated. Conclusion: Ripk3 signaling plays distinct roles in HSCs in response to different doses of IR. High-dose IR induces Ripk3-dependent NF-κB/survival signaling, which is required for the survival of HSCs which fail to repair the damage. Thus temporal inhibition of Ripk3-NF-κB signaling might help to remove the damaged HSCs thus preventing the occurrence of LT-RD. However multiple low-doses of IR induces Ripk3 activation in HSCs which represses leukemia development by inducing both ROS-p38-p16-mediated senescence and Ripk3-Mlkl-mediated necroptosis. Induced activation of Mlkl-necroptosis might help to repress leukemia development by removing damaged HSCs. 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: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Rockefeller University Press ; 2014
    In:  Journal of Experimental Medicine Vol. 211, No. 6 ( 2014-06-02), p. 1093-1108
    In: Journal of Experimental Medicine, Rockefeller University Press, Vol. 211, No. 6 ( 2014-06-02), p. 1093-1108
    Abstract: Leukemic stem cells (LSCs) isolated from acute myeloid leukemia (AML) patients are more sensitive to nuclear factor κB (NF-κB) inhibition-induced cell death when compared with hematopoietic stem and progenitor cells (HSPCs) in in vitro culture. However, inadequate anti-leukemic activity of NF-κB inhibition in vivo suggests the presence of additional survival/proliferative signals that can compensate for NF-κB inhibition. AML subtypes M3, M4, and M5 cells produce endogenous tumor necrosis factor α (TNF). Although stimulating HSPC with TNF promotes necroptosis and apoptosis, similar treatment with AML cells (leukemic cells, LCs) results in an increase in survival and proliferation. We determined that TNF stimulation drives the JNK–AP1 pathway in a manner parallel to NF-κB, leading to the up-regulation of anti-apoptotic genes in LC. We found that we can significantly sensitize LC to NF-κB inhibitor treatment by blocking the TNF–JNK–AP1 signaling pathway. Our data suggest that co-inhibition of both TNF–JNK–AP1 and NF-κB signals may provide a more comprehensive treatment paradigm for AML patients with TNF-expressing LC.
    Type of Medium: Online Resource
    ISSN: 1540-9538 , 0022-1007
    RVK:
    Language: English
    Publisher: Rockefeller University Press
    Publication Date: 2014
    detail.hit.zdb_id: 218343-2
    detail.hit.zdb_id: 1477240-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, ( 2020-02-07)
    Abstract: c-Myc (Myc hereafter) is found to be deregulated and/or amplified in most acute myeloid leukemias (AML). Almost all AML cells are dependent upon Myc for their proliferation and survival. Thus Myc has been proposed as a critical anti-AML target. Myc has Max-mediated trans-activational and Miz1-mediated trans-repressional activities. The role of Myc-Max-mediated trans-activation in the pathogenesis of AML has been well-studied; however the role of Myc-Miz1-mediated trans-repression in AML is still somewhat obscure. MycV394D is a mutant form of Myc which lacks trans-repressional activity due to a defect in its ability to interact with Miz1. We found that, compared to Myc, the oncogenic function of MycV394D is significantly impaired. The AML/myeloproliferative disorder which develops in mice receiving MycV394D-transduced hematopoietic stem/progenitor cells (HSPCs) is significantly delayed compared to mice receiving Myc-transduced HSPCs. Using a murine MLL-AF9 AML model, we found that AML cells expressing MycV394D (intrinsic Myc deleted) are partially differentiated and show reductions in both colony-forming ability in vitro and leukemogenic capacity in vivo. The reduced frequency of leukemia stem cells (LSCs) among MycV394D-AML cells and their reduced leukemogenic capacity during serial transplantation suggest that Myc-Miz1 interaction is required for the self-renewal of LSCs. In addition, we found that MycV394D-AML cells are more sensitive to chemotherapy than are Myc-AML cells. Mechanistically, we found that the Myc represses Miz1-mediated expression of Cebpα and Cebpδ, thus playing an important role in the pathogenesis of AML by maintaining the undifferentiated state and self-renewal capacity of LSCs.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    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: Stem Cells and Development, Mary Ann Liebert Inc, Vol. 28, No. 22 ( 2019-11-15), p. 1486-1497
    Type of Medium: Online Resource
    ISSN: 1547-3287 , 1557-8534
    Language: English
    Publisher: Mary Ann Liebert Inc
    Publication Date: 2019
    detail.hit.zdb_id: 2142305-2
    detail.hit.zdb_id: 2142214-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3752-3752
    Abstract: Background: In addition to inducing apoptosis, tumor necrosis factor-α (TNFα) and FAS-induced death receptor signaling also induce necroptosis, a recently defined type of programed necrosis, through activation of the RIP1/RIP3-kinase pathway. The role of death receptor signaling-induced apoptosis in the development of leukemia has been well documented. However, the role of necroptotic signaling in the pathogenesis of leukemia has not been studied. Methods: RIP1/RIP3 signaling was inactivated in murine and human acute myeloid leukemia (AML) cells by genetic deletion of RIP1/RIP3 or pharmacologic inhibition using the specific inhibitor Necrostatin-1. The effects of RIP1/RIP3 signal inactivation on the behavior and interferon-γ (IFNγ)-induced differentiation of AML cells were evaluated by morphologic analysis, cell surface markers, in vitro colony-forming ability and in vivo transplantation/leukemogenic capacity. The responses of mice transplanted with Rip1-/- or Rip3-/-murine AML to IFNγ treatment were compared to the mice transplanted with wild-type AML. Results: We found that most types of AML cells produce TNFα and already show basal level activation of RIP1/RIP3 signaling but they do not undergo necroptosis. Using both pharmacological inhibition and genetic deletion techniques, we determined that inactivation of the RIP1/RIP3 signal resulted in partial differentiation of both human AML cell lines and MLL-AF9-transduced murine AML cells as demonstrated by studies of morphology, cell surface markers and colony-forming ability. Rip1 or Rip3 deletion significantly repressed the leukemogenic capacity of murine AML cells in vivo. In addition, inactivation of RIP1/RIP3 signaling significantly enhanced the complete differentiation of AML cells induced by IFNγ. The combination of RIP1/RIP3 inactivation and IFN treatment significantly further attenuated the clonogenic capacity of both primary AML cells and AML cell lines. Such combination treatment also further compromised the leukemogenic ability of AML cells in vivo. Mechanistically, we found that the RIP1/RIP3 pathway regulates the expression of SOCS1, a key negative regulator of IFNγ signaling. Inactivation of RIP1/RIP3 signaling in AML cells renders them hypersensitive to IFNγ -induced monocytic lineage maturation. Conclusion: Resistance to apoptosis is one of the key mechanisms involved in the development of drug resistance in leukemic cells. RIP1/RIP3-mediated necroptosis induced by TNFα has been proposed to be an alternative therapeutic strategy to treat leukemia. However, our studies suggested that inhibition of RIP1/RIP3-mediated necroptotic signaling might be useful strategy for AML treatment when combined with IFNγ. 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 ...
  • 10
    In: Molecular Oncology, Wiley, Vol. 7, No. 6 ( 2013-12), p. 1069-1082
    Abstract: p27 is specifically expressed in a small subset of MLL‐AF9 LSC but not in HOXA9/MEIS1 LSC. The niche‐generated factors Flt3L and SCF induce p27 expression in MLL‐AF9 LSC but not in HOXA9/MEIS1 LSC. p27 restricts proliferation and maintains resistance to chemotherapeutic agents in MLL‐AF9 LSC. Over‐expression of p27 in HOXA9/MEIS1 LSC leads to quiescence and resistance to chemotherapeutic agents.
    Type of Medium: Online Resource
    ISSN: 1574-7891 , 1878-0261
    Language: English
    Publisher: Wiley
    Publication Date: 2013
    detail.hit.zdb_id: 2415106-3
    detail.hit.zdb_id: 2322586-5
    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...