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
    Online-Ressource
    Online-Ressource
    Elsevier BV ; 2007
    In:  Protein Expression and Purification Vol. 56, No. 2 ( 2007-12), p. 220-228
    In: Protein Expression and Purification, Elsevier BV, Vol. 56, No. 2 ( 2007-12), p. 220-228
    Materialart: Online-Ressource
    ISSN: 1046-5928
    Sprache: Englisch
    Verlag: Elsevier BV
    Publikationsdatum: 2007
    ZDB Id: 1471688-4
    SSG: 12
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    Online-Ressource
    Online-Ressource
    Impact Journals, LLC ; 2014
    In:  Oncotarget Vol. 5, No. 14 ( 2014-07-30), p. 5559-5569
    In: Oncotarget, Impact Journals, LLC, Vol. 5, No. 14 ( 2014-07-30), p. 5559-5569
    Materialart: Online-Ressource
    ISSN: 1949-2553
    URL: Issue
    Sprache: Englisch
    Verlag: Impact Journals, LLC
    Publikationsdatum: 2014
    ZDB Id: 2560162-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 2323-2323
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 2323-2323
    Kurzfassung: Melanoma is a lethal form of cutaneous cancer which accounts for 80% of skin cancer deaths owing to its highly metastatic behavior and few clinically effective therapies. More than 60% of melanoma patients present with mutations in B-Raf, a serine/threonine kinase that increases proliferation and survival through the constitutive activation of ERK signaling. Inhibition of mutated B-Raf with specific inhibitors like vemurafenib reduces tumor growth and extends patient survival. However, clinical responsiveness to vemurafenib is limited in most melanoma patients, prompting analysis of additional pathways that could be targeted to extend vemurafenib responsiveness or overcome resistance. Towards this goal, we examined the impact of vemurafenib on substrates or cascades activated in B-Raf mutant and wild-type melanoma cells. We were particularly interested in assessing the impact of mutant B-Raf inhibition on deubiquitinases (DUBs), which are overexpressed or activated in many tumor types. DUBs control the stability, activity and localization of multiple proteins and are emerging as therapeutic targets. We found that vemurafenib treatment or lentiviral-mediated B-Raf knockdown reduced the activity of several DUBs in B-Raf mutant melanoma cells, including Usp9x, Usp14 and Usp5. These DUBs were not affected by vemurafenib in w/t B-Raf-expressing cells. Interestingly lentiviral-mediated knockdown of Usp5, but not Usp9x, reduced melanoma growth and increased vemurafenib sensitivity whereas Usp5 overexpression suppressed vemurafenib activity. Inhibition of Usp5 activity by vemurafenib or by Usp5 shRNA KD alone had little effect on melanoma cell apoptosis. However B-Raf inhibition in Usp5 KD cells resulted in & gt;5-fold increased melanoma cell apoptosis. Further, Usp5 KD increased accumulation of p53 and p53-related proteins as well as their transcriptional targets. Our data clearly shows that mutant B-Raf activates Usp5 to suppress apoptotic responsiveness to vemurafenib and kinase inhibition alone was not sufficient to completely suppress Usp5 activity. Silencing studies demonstrate that Usp5 inhibition will enhance cell killing in response to B-Raf inhibition in melanoma. To address that potential, we compared anti-tumor activity in mice treated with vemurafenib and a small molecule DUB inhibitor, WP1130, with activity against Usp5. Treatment with either agent alone reduced tumor growth while mice receiving both vemurafenib and WP1130 had enhanced anti-tumor responses. Together, these results suggest that mutant B-Raf activates Usp5 activity to suppress p53 induction and apoptotic responsiveness to vemurafenib. These observations also suggest that further testing and refinement of Usp5 inhibitors as an additional approach to targeted therapy for melanoma are warranted. Citation Format: Harish Potu, Luke F. Peterson, Monique Verhaegen, Moshe Talpaz, Nicholas J. Donato. Mutant B-Raf-mediated activation of deubiquitinases in melanoma defines Usp5 as a potential therapeutic target. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2323. doi:10.1158/1538-7445.AM2013-2323
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2013
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 4435-4435
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 4435-4435
    Kurzfassung: Melanoma is an aggressive skin cancer characterized by gene mutations, high metastatic potential and few clinically effective systemic therapies. BRAF, MEK1 kinase inhibitors and CTLA4 antagonist have improved patient survival, but only a subset of patients respond or derived long-lasting clinical benefit, illustrating the need for discovery of additional targets and pathways that may be inhibited with small molecules. Given the clinical impact of inhibitors of the ubiquitin-proteasome system in other cancers, we explored the potential for aberrant deubiquitinase (DUB) activity to play a role in the tumorigenic properties of melanoma. We also assessed whether specific DUBs play a role in apoptotic responsiveness and resistance to kinase inhibitors. Initially, we noted that the activity of several DUBs, including Usp9x and Usp5, was elevated in melanoma compared to normal melanocytes. Further, Usp9x and Usp5 expression and activity were up-regulated by expression of BRAFV600E in 293T cells and kinase inhibition partially suppressed both Usp9x and Usp5 activity in vemurafenib sensitive, but not resistant cells. Usp9x knockdown (KD) in BRAF mutant or wild-type melanoma did not affect 2-dimensional cell growth or survival but significantly amplified the apoptotic activity of BRAF or MEK1 inhibitors, respectively. Usp5 KD in melanoma suppressed cell growth by reinforced the S/G2-M checkpoint, enhanced extrinsic caspase activation through modulation of p53 and FAS levels and amplified the apoptotic activity of kinase inhibitors (and other chemotherapeutic agents) and overcame vemurafenib resistance in A375R cells. To further assess the clinical relevance of activated DUBs in melanoma, primary human melanoma explants characterized for their metastatic efficiency in patients and NSG mice demonstrated that Usp9x and Usp5 expression and activity were elevated in efficient metastasizers, but only a subset of inefficient metastasizers, suggesting a potential role for DUBs in the regulation of metastatic potential. To investigate that possibly, control and Usp9x KD cells were grown in Matrigel or suspension culture and colony growth and survival, as well as Usp9x activity were examined. Usp9x KD significantly suppressed melanoma colony growth and fully reduced survival of highly metastatic SK-Mel147 cells in 3D culture. Growth in 3D conditions resulted in activation of Usp9x, but also increased their sensitivity (IC50 & lt;400 nM) to our partially selective Usp9x inhibitor, EOAI3402143 (G9). G9 inhibits both Usp9x and Usp5 activity and phenocopied the activities of both Usp9x and Usp5 KD cells. G9 was safely administered to NSG mice, with complete block of melanoma growth. We conclude that targeted inhibition of specific DUBs will suppress metastatic melanoma and amplify existing melanoma therapies. Citation Format: Harish Potu, Anupama Pal, Hanshi Sun, Luke Peterson, Moshe Talpaz, Monique Verhaegen, Juxiang Cao, Ugur Eskiocak, Sean Morrison, Nicholas J. Donato. Deubiquitinases Usp9x and Usp5 control tumorigenicity and apoptotic responsiveness in malignant melanoma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4435. doi:10.1158/1538-7445.AM2014-4435
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2014
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    In: Blood, American Society of Hematology, Vol. 125, No. 23 ( 2015-06-04), p. 3588-3597
    Kurzfassung: Deubiquitinases Usp9x and Usp24 regulate Mcl-1 and myeloma cell survival. Small-molecule–mediated Usp9x/Usp24 inhibition induces apoptosis and blocks myeloma tumor growth in vivo.
    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 ...
  • 6
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 8, No. 1 ( 2017-02-15)
    Kurzfassung: ETS transcription factors are commonly deregulated in cancer by chromosomal translocation, overexpression or post-translational modification to induce gene expression programs essential in tumorigenicity. Targeted destruction of these proteins may have therapeutic impact. Here we report that Ets-1 destruction is regulated by the deubiquitinating enzyme, Usp9x, and has major impact on the tumorigenic program of metastatic melanoma. Ets-1 deubiquitination blocks its proteasomal destruction and enhances tumorigenicity, which could be reversed by Usp9x knockdown or inhibition. Usp9x and Ets-1 levels are coincidently elevated in melanoma with highest levels detected in metastatic tumours versus normal skin or benign skin lesions. Notably, Ets-1 is induced by BRAF or MEK kinase inhibition, resulting in increased NRAS expression, which could be blocked by inactivation of Usp9x and therapeutic combination of Usp9x and MEK inhibitor fully suppressed melanoma growth. Thus, Usp9x modulates the Ets-1/NRAS regulatory network and may have biologic and therapeutic implications.
    Materialart: Online-Ressource
    ISSN: 2041-1723
    Sprache: Englisch
    Verlag: Springer Science and Business Media LLC
    Publikationsdatum: 2017
    ZDB Id: 2553671-0
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    Online-Ressource
    Online-Ressource
    American Society of Hematology ; 2019
    In:  Blood Vol. 134, No. Supplement_1 ( 2019-11-13), p. 3936-3936
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 3936-3936
    Kurzfassung: Bromodomain and Extra Terminal (BET) proteins which include ubiquitously expressed BRD2, BRD3, BRD4, and testis-specific BRDT, are epigenetic "readers" and play a major role in epigenetic regulation of gene transcription. BET family proteins are crucial for acute myeloid leukemia (AML) maintenance and expression of BRD4 is a poor prognostic indicator in AML. BET inhibitor activity is associated with repression of oncogenic c-MYC and their efficacy in pre-clinical models of cancer has led to rapid development of BET inhibitors entering clinical trials. Depletion of key oncogenic proteins in tumor cells could achieve much better clinical efficacy than inhibition of the same proteins. Therefore, we have developed novel and highly potent BET degrader, QCA570, based on the proteolysis targeting chimera (PROTAC) concept. This "BET-degrader" is based upon a new class of BET inhibitor, HYD276. QCA570 effectively degrades all BET-BRD proteins at concentrations as low as 30 pM within a few hours of treatment in the RS4;11 leukemia cell line and achieves IC50 values of 50 pM in inhibition of RS4;11 cell growth. We tested QCA570 (BET-degrader) in a panel of 102 primary AML samples in ex-vivo cultures. Leukemia stem cells (LSC) were enriched by positive selection with CD34 antibody. Cells were plated in LSC conducive medium containing growth factors. Cell death and apoptosis induction was measured by flow-cytometry for DAPI-Annexin V binding after incubation with BET-inhibitor and BET-degrader for 3 days in the presence of drug. The AML cells were obtained from newly diagnosed AML (N=87) or relapse/refractory AML (N=15). The median IC50s was 120 pM (range 32 pm to 23 nM). Only 13 of 102 AML samples had an IC50 〉 1 nM with the BET degrader. In contrast median IC50 for the inhibitor HYD276 was 30,000 fold higher at 3.5 µM (range 8 nM to 151 µM). An IC50 of greater than 10 µM HYD276 was considered as resistant to BET-inhibitor. BET-inhibitor resistant CD34 enriched LSCs were sensitive to the degrader QCA570, irrespective of mutation status and chromosomal aberrations, suggesting that there are additional mechanisms of action of the degrader as compared to the inhibitor. The AML cells were heterogeneous for the expression of mutant FLT-3 (36%), NPM1 (22%), IDH1/2 (16%), CEBPalpha (2%), DNMT3A (2%) and other mutations (10%). Based on classical karyotype 30% of them had a normal karyotype and 55% had abnormal karyotype (15% data missing). The degradation of BRD4 and repression of c-MYC was confirmed by immunoblotting after a 4 hr exposure of LSCs to degrader. Moreover, treatment of normal CD34 positive hematopoietic cells in apoptosis assay and colony formation assays suggests lack of toxicity to normal hematopoiesis. In conclusion, QCA570 represents a highly potent and efficacious panBET-BRD degrader undergoing extensive pre-clinical evaluation for the treatment of acute leukemias. Disclosures Peterson: Penrose TherapeuTx: Employment. Wang:Oncopia Therapeutics: Consultancy, Equity Ownership, Patents & Royalties. Talpaz:Samus Therapeutics: Research Funding; Imago BioSciences: Consultancy, Research Funding; Celgene: Consultancy, Research Funding; CTI BioPharma: Research Funding; Constellation: Research Funding; Incyte: Research Funding; Novartis: Research Funding.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2019
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 8
    Online-Ressource
    Online-Ressource
    Springer Science and Business Media LLC ; 2011
    In:  Apoptosis Vol. 16, No. 12 ( 2011-12), p. 1229-1244
    In: Apoptosis, Springer Science and Business Media LLC, Vol. 16, No. 12 ( 2011-12), p. 1229-1244
    Materialart: Online-Ressource
    ISSN: 1360-8185 , 1573-675X
    Sprache: Englisch
    Verlag: Springer Science and Business Media LLC
    Publikationsdatum: 2011
    ZDB Id: 1495863-6
    SSG: 12
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 2881-2881
    Kurzfassung: 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.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2016
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 10
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1748-1748
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1748-1748
    Kurzfassung: Usp9x regulates tumor cell survival and responsiveness to targeted-, chemo- and radio-therapy in a broad number of tumors through effects on multiple proteins. The role of Usp9x in human pancreatic cancer is largely unknown, although a recent study demonstrated that Usp9x co-operated in promoting KrasG12D tumorigenesis in mice. To examine Usp9x tumorigenicity and therapeutic potential of Usp9x inhibition in human pancreatic cancer vs. mouse pancreatic tumor models we developed 3D cultures of cell lines established from constitutive (8041) and doxycycline-inducible (4668) KrasG12D/Tp53R172H mouse pancreatic tumors, established human cell lines (BxPC3, PANC1 and MIAPACA2) and spontaneously immortalized human pancreatic patient tumor derived cell lines (UM2, UM6, UM16 and UM76). The effect of Usp9x knockdown (KD), overexpression (OE) or Usp9x activity inhibition by our small molecule deubiquitinase inhibitor EOAI3402143 (G9) were assessed by growth in 2D and 3D culture and anti-tumor efficacy studies in mouse (8041) and human (MIAPACA2) xenograft models. Usp9x KD in constitutive KrasG12D/Tp53R172H 8041 cells led to a 3-fold increase in the number of colonies formed in 3D and Usp9x KD sustained 3D colony growth even after withdrawal of mutant Kras support in KrasG12D/Tp53R172H inducible 4668 cells. Usp9x-OE in 8041 cells decreased 3D colony growth by 2.5-fold. These results contrast with outcomes in human pancreatic tumors where Usp9x KD induced either rapid apoptosis (in MIAPACA2) or reduced 3D colony formation by & gt;50% (in PANC1). Usp9x-OE in PANC1 cells enhanced 3D colony growth (2-fold) and increased invasion activity in Boyden chambers (3.5-fold). Mechanistically, we found differential effects of Usp9x on mutant Kras protein expression levels in mouse vs human cells. In vivo G9 treatment (15 mg/kg, every other day) of mouse 8041 xenografts showed a statistically insignificant trend towards tumor growth inhibition but effectively inhibited MIAPACA2 tumor growth in mice. Usp9x KD in human pancreatic patient tumor cell lines inhibited 3D colony growth by & gt;75% and this activity was phenocopied by G9 with nM efficacy. We conclude that Usp9x acts as a tumor promoter in established human pancreatic cancer and a tumor suppressor in murine cells from genetically engineered pancreatic tumors, possibly through differential regulation of mutant Kras. Overall, these results suggest that Usp9x may be a novel therapeutic target in pancreatic cancer. Note: This abstract was not presented at the meeting. Citation Format: Anupama Pal, Marina Pasca Di Magliano, Diane Simeone, Luke Peterson, Harish Potu, Moshe Talpaz, Nicholas Donato. Usp9x as a novel therapeutic target in human pancreatic cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1748. doi:10.1158/1538-7445.AM2015-1748
    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 ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...