Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
Type of Medium
Language
Subjects(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 2881-2881
    Abstract: Transcription factors are frequently deregulated in cancer cells and may be good therapeutic targets, but few successful targeting strategies have been reported. Deubiquitinases (DUBs) are specialized enzymes that regulate the ubiquitin (Ub) content on many proteins and DUB expression and activity are elevated in a number of cancers where they can act to alter tumor suppressor and/or oncoprotein levels. We previously described Usp9x activity and expression in melanoma; here we sought to investigate its role in primary melanoma and metastatic disease. Usp9x was upregulated in tumor cells compared to normal melanocytes and Usp9x expression and activity were found to be essential for 3D growth and melanoma tumor expansion in vivo. We defined the Usp9x ubiquitinated protein landscape and demonstrate that Usp9x regulates Ets-1, a cancer-promoting transcription factor. Usp9x binds, deubiquitinates and thereby stabilizes Ets-1 protein, and primary tissue and tumor analysis demonstrated elevated and coincident Usp9x/Ets-1 protein expression in melanoma compared to normal skin or benign nevi. Usp9x knockdown or Usp9x inhibition with small molecule G9 reduced Ets-1 protein levels and blocked tumor growth in vitro and in vivo. Conversely, Usp9x overexpression in melanoma cells increased Ets-1 protein levels and enhanced 3D tumor growth in vitro and in vivo, which were all reversible by treatment with G9. We conclude that Usp9x is essential for Ets-1 protein stability and may be therapeutically exploited with small molecule Usp9x inhibitors to reduce Ets-1-dependent gene expression and tumorigenicity. Citation Format: Harish Potu, Luke F. Peterson Peterson, Malathi Kandarpa Kandarpa, Anupama Pal Pal, Hanshi Sun Sun, Paul W. Harms Harms, Peter C. Hollenhorst Hollenhorst, Ugur Eskiocak Eskiocak, Moshe Talpaz Talpaz, Nicholas J. Donato Donato. Deubiquitinase Usp9x controls tumorigenicity through regulation of the Ets-1 transcription factor in melanoma. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2881.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Clinical Immunology, Elsevier BV, Vol. 150, No. 2 ( 2014-02), p. 201-209
    Type of Medium: Online Resource
    ISSN: 1521-6616
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2014
    detail.hit.zdb_id: 1462862-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Cold Spring Harbor Laboratory ; 2003
    In:  Genes & Development Vol. 17, No. 4 ( 2003-02-15), p. 455-460
    In: Genes & Development, Cold Spring Harbor Laboratory, Vol. 17, No. 4 ( 2003-02-15), p. 455-460
    Abstract: ISG15 is one of the most strongly induced genes upon viral infection, type I interferon (IFN) stimulation, and lipopolysaccharide (LPS) stimulation. Here we report that mice lacking UBP43, a protease that removes ISG15 from ISGylated proteins, are hypersensitive to type I IFN. Most importantly, in UBP43-deficient cells, IFN-β induces a prolonged Stat1 tyrosine phosphorylation, DNA binding, and IFN-mediated gene activation. Furthermore, restoration of ISG15 conjugation in protein ISGylation-defective K562 cells increases IFN-stimulated promoter activity. These findings identify UBP43 as a novel negative regulator of IFN signaling and suggest the involvement of protein ISGylation in the regulation of the JAK-STAT pathway.
    Type of Medium: Online Resource
    ISSN: 0890-9369 , 1549-5477
    RVK:
    Language: English
    Publisher: Cold Spring Harbor Laboratory
    Publication Date: 2003
    detail.hit.zdb_id: 1467414-2
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2004
    In:  Oncogene Vol. 23, No. 24 ( 2004-05-24), p. 4255-4262
    In: Oncogene, Springer Science and Business Media LLC, Vol. 23, No. 24 ( 2004-05-24), p. 4255-4262
    Type of Medium: Online Resource
    ISSN: 0950-9232 , 1476-5594
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2004
    detail.hit.zdb_id: 2008404-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 110, No. 3 ( 2007-08-01), p. 799-805
    Abstract: Nonrandom and somatically acquired chromosomal translocations can be identified in nearly 50% of human acute myeloid leukemias. One common chromosomal translocation in this disease is the 8q22;21q22 translocation. It involves the AML1 (RUNX1) gene on chromosome 21 and the ETO (MTG8, RUNX1T1) gene on chromosome 8 generating the AML1-ETO fusion proteins. In this review, we survey recent advances made involving secondary mutational events and alternative t(8;21) transcripts in relation to understanding AML1-ETO leukemogenesis.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Society of Hematology ; 2005
    In:  Blood Vol. 106, No. 11 ( 2005-11-16), p. 2721-2721
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 2721-2721
    Abstract: The transcription factor AML1 (RUNX1) plays a critical role in normal hematopoiesis and in the development of leukemia. The phenotype generated by knockout of the AML1 gene demonstrates that AML1 is necessary for normal development of fetal hematopoietic cells. Conditional knockout of AML1 expression in adult bone marrow leads to lineage-specific effects on B- and T-cell maturation and pronounced inhibition of common lymphocyte progenitor production. In addition, the AML1-deficient adult mice exhibited inefficient platelet production and a mild myeloproliferative phenotype characterized by an increase in peripheral blood neutrophils, an increase in myeloid progenitor populations, and extramedullary hematopoiesis composed of maturing myeloid and erythroid elements. Previous studies have shown that phosphorylation of AML1, particularly at serines 276 and 303, can affect transcriptional activation. We have now shown that phosphorylation of AML1 serines 276 and 303 can be blocked in vivo by inhibitors of the cyclin-dependent kinases (Cdks) Cdk1 and Cdk2. In addition, these two sites can be phosphorylated in vitro by purified, active Cdk1/cyclin B and Cdk2/cyclin A, but not by Cdk4/cyclin D. Mutation of AML1 serines 276 and 303 reduces AML1 activity during most phases of the cell cycle. In addition, mutation of serines 276 and 303 prevents phosphorylation from occurring at two nearby sites (serine 293 and threonine 300). Mutation of serine 293 and threonine 300 causes an increase of AML1 activity, especially in cells arrested in G2/M by treatment with nocodazole. Since Cdk phosphorylation can be stimulatory (serines 276 and 303) or inhibitory (serine 293, threonine 300), the interplay of Cdk phosphorylation at different sites might confer a very fine level of regulation of AML1 activity.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3005-3005
    Abstract: Abstract 3005 Multiple myeloma (MM) patients are typically treated with a number of combinations of unconventional drugs that provide excellent and sometimes durable responses. However, most patients are expected to relapse or progress and relapsed disease is usually less sensitive to chemotherapy. Therefore, drug resistance remains a key concern in MM therapy. Some important clues regarding MM drug and apoptotic resistance have been obtained from biochemical and molecular studies of pro-survival genes. Mcl-1 is one member of the Bcl-2 family with strong anti-apoptotic activity. Mcl-1 binds pro-apoptotic proteins such as Bim and Bak, forming hetero-dimers to suppress apoptosis. Specific knockdown of Mcl-1 but not other anti-apoptotic Bcl-2 proteins (Bcl-2, Bcl-xL) induces apoptosis in MM cells and a critical level of Mcl-1 is essential to MM cell viability. Also, drug resistant MM patients are reported to express higher levels of Mcl-1 when compared to drug naïve patients. These observations suggest that Mcl-1 plays a key role in MM drug and apoptotic resistance. Mcl-1 is unique among survival proteins as its cellular protein level is primarily controlled by its ubiquitination and proteasomal degradation. Recently Usp9x, a high MW de-ubiquitinase (DUB), was shown to bind and stabilize Mcl-1 by mediating its de-ubiquitination and loss of recognition by the proteasome. Usp9x was also shown to be highly expressed in MM patients with short term progression-free survival. Analysis of Usp9x gene expression in MM patients treated at our center supports a correlation between elevated Usp9x expression ( 〉 4-fold over normal PBMCs) and poor patient prognosis. These observations suggest that agents that inhibit Usp9x activity may be effective in controlling Mcl-1 levels and have therapeutic impact in MM patients. Here we describe a small molecule Usp9x inhibitor, WP1130, that causes a rapid reduction in Mcl-1 protein levels and stimulates apoptosis in MM cells. We further demonstrate that the anti-tumor activity of WP1130 is mediated through inhibition of Usp9x and other active DUBs in B-cell tumors. WP1130-mediated inhibition of Usp9x activity promotes Mcl-1 ubiquitination leading to its rapid (1-4 hours) proteasomal degradation with subsequent induction of apoptosis. Pretreatment of MM cells with proteasome inhibitors (MG-132 or bortezomib) prior to WP1130 treatment blocked the down-regulation of Mcl-1, demonstrating proteasome involvement in regulation of Mcl-1 levels by WP1130. Primary MM tumor cells (bone marrow aspirates) derived from newly diagnosed and advanced therapy refractory patients were also screened for their responsiveness to WP1130 by DUB activity assays. Samples were obtained from a newly diagnosed plasmacytoma/plasma cell leukemia patient and 4 MM patients refractory to the Velcade/Doxorubicin/Dexamethasone (VDD) regimen. Usp9x activity was detected in 4 of 5 patient specimens and WP1130 treatment reduced Mcl-1 protein levels and induced apoptosis only in samples with active Usp9x. Furthermore, WP1130 completely eliminated CD138+ cells after 24 hours in MM patient specimens with active Usp9x while only an 11% reduction was measured in MM cells that did not exhibit Usp9x activity. Cytogenetic prognostic predictors (del13) did not distinguish between WP1130-responsive and non-responsive cells. Our results suggest that inhibition of Usp9x activity by WP1130 is associated with Mcl-1 down-regulation in MM cells. These results also suggest that drug-refractory MM patients with elevated Usp9x activity would likely benefit from Usp9x inhibitor-based therapy. Preliminary MM tumor studies have shown that WP1130 suppresses tumor growth in vivo and can be administered safely. Together, these results suggest that WP1130-mediated Usp9x inhibition may be an effective and novel approach in treating progressive or drug-refractory MM patients. Disclosures: Jakubowiak: Millenium, Celgene, Bristol-Myers Squibb, Johnson & Johnson Ortho-Centocor: Honoraria; Millennium, Bristol-Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Millennium, Celgene, Centocor-Ortho Biotech: Speakers Bureau.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 3336-3336
    Abstract: t(8;21) is one of the most common chromosomal abnormalities associated with acute myeloid leukemia (AML). The resulting AML1-ETO fusion protein is directly involved in the pathogenesis of AML but is not able to cause leukemia on its own. We have shown previously that a C-terminal splice variant of t(8;21), AML1-ETO9a (AE9a), is sufficient to promote AML in mice. Here we show that the leukemia-initiating cells reside in the Lin-/Sca1-/cKit+ population. To identify molecular targets directly modulated by AE9a, we compared the gene expression profile to the promoter occupancy (ChIP-on-chip) profile of this population. We identified 1485 genes deregulated by AE9a, among which 544 genes are also ChIP-on-chip targets. CD45, a negative regulator of cytokine/growth factor receptor and JAK/STAT signaling, was greatly down-regulated in AE9a leukemia cells. We further show that t(8;21) AML-M2 patient samples have lower CD45 levels compared to non-t(8;21) AML-M2 samples. Interestingly, JAK1 and JAK2 were upregulated by AE9a and both CD45 and JAK1 are ChIP-on-chip targets. In addition we show that JAK/STAT signaling was enhanced in the leukemia cells. Consequently, the leukemia cells are more susceptible to JAK2 inhibitors than wild type cells. Our results indicate that AE9a enhances JAK/STAT signaling by directly modulating regulators of this signaling pathway, which provides a potential novel approach to treating t(8;21) AML.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Society of Hematology ; 2008
    In:  Blood Vol. 112, No. 11 ( 2008-11-16), p. 3213-3213
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 3213-3213
    Abstract: In Chronic Myeloid Leukemia (CML) the 9;22 chromosomal translocation and corresponding BCR-ABL protein are present in the most primitive hematopoietic stem/progenitors (Lin−/CD38−/CD34+). These cells are refractory to the effect of BCR-ABL tyrosine kinase inhibitors. The mechanism of this resistance has not been fully elucidated but is clearly distinct from the mechanism of resistance in the more mature CML cells. The p53 gene is rarely mutated in the chronic phase of CML. BCR-ABL is able to positively affect p53 expression whose potential proapoptotic effect may be balanced by survival signals such as Bcl-XL and Stat signaling. However, BCR-ABL also positively regulates HDM2, the negative regulator of p53, which may be the alternative mechanism of counteracting the induced p53. In an effort to facilitate a cytotoxic effect directed against the refractory CML primitive stem/progenitor cells we elected to explore the role of stabilizing the p53 protein. Accordingly we tested a novel inhibitor of the HDM2-p53 interaction (MI-219; Ascenta), which interferes with unmutated p53 degradation. MI- 219 induced reproducible cytotoxicity in four CML blast-crisis cell lines with intact p53 (WDT2, WDT3, BV173 and BV173R) with an IC50 ~2 microM. The BV173R cell line which has the Imatinib resistant T315I mutation displayed a cytotoxic effect with the MI- 219 equal to its parental BV173 cell line (IC50 ~2 microM). Responses were associated with the induction of p53 protein, its targets p21WAF1 and PUMA, and cleavage of PARP. The K562 cell line with mutated p53 did not respond to MI-219 as expected. MI-219 had a modest cytotoxic effect on magnetically separated (MACS) CD34+ cells from CML patients as a single agent (range of 30–50% cell death at 5 microM MI-219). Nevertheless, MI-219 markedly enhanced the cytotoxic effect of Imatinib on CD34+ cells, while as a single agent Imatinib induced 15–30% apoptosis. However the combination of 2 microM Imatinib and 5 microM MI-219 led to a cytotoxic effect averaging 76.4 ± 10.6% apoptosis. This enhanced cytotoxic effect was further noted in flow cytometrically sorted progenitor (Lin−/CD38+/CD34+) populations (~86.7% apoptosis). This combination equally induced apoptosis in primitive progenitor/stem cells (Lin−/CD38−/CD34+; ~83.0%), despite the minimal affect of each agent when given alone (Imatinib, ~20.8 % apoptosis; MI-219, ~36.9% apoptosis). This cytotoxic effect in primary CML cells was again associated with the induction of p53, p21WAF1, and the cleavage of PARP. Here we demonstrate that an increased level of p53 bypasses T315I associated resistance to Imatinib, and in combination with Imatinib generates a substantial cytotoxic effect in early progenitors, which are otherwise refractory to the effect of either agent alone. Thus these observations propose that the combination of MI-219 an HDM2-inhibitor with Imatinib may facilitate the eradication of minimal residual disease present within the primitive Lin−/CD38−/CD34+ population of CML.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 135, No. 18 ( 2020-04-30), p. 1560-1573
    Abstract: Expression of the cell cycle regulatory gene CDK6 is required for Philadelphia-positive (Ph+) acute lymphoblastic leukemia (ALL) cell growth, whereas expression of the closely related CDK4 protein is dispensable. Moreover, CDK6 silencing is more effective than treatment with the dual CDK4/6 inhibitor palbociclib in suppressing Ph+ ALL in mice, suggesting that the growth-promoting effects of CDK6 are, in part, kinase-independent in Ph+ ALL. Accordingly, we developed CDK4/6–targeted proteolysis-targeting chimeras (PROTACs) that inhibit CDK6 enzymatic activity in vitro, promote the rapid and preferential degradation of CDK6 over CDK4 in Ph+ ALL cells, and markedly suppress S-phase cells concomitant with inhibition of CDK6-regulated phospho-RB and FOXM1 expression. No such effects were observed in CD34+ normal hematopoietic progenitors, although CDK6 was efficiently degraded. Treatment with the CDK6-degrading PROTAC YX-2-107 markedly suppressed leukemia burden in mice injected with de novo or tyrosine kinase inhibitor–resistant primary Ph+ ALL cells, and this effect was comparable or superior to that of the CDK4/6 enzymatic inhibitor palbociclib. These studies provide “proof of principle” that targeting CDK6 with PROTACs that inhibit its enzymatic activity and promote its degradation represents an effective strategy to exploit the “CDK6 dependence” of Ph+ ALL and, perhaps, of other hematologic malignancies. Moreover, they suggest that treatment of Ph+ ALL with CDK6-selective PROTACs would spare a high proportion of normal hematopoietic progenitors, preventing the neutropenia induced by treatment with dual CDK4/6 inhibitors.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages