Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 23, No. 20 ( 2017-10-15), p. 6227-6238
    Abstract: Purpose: Myxoid liposarcoma is an aggressive disease with particular propensity to develop hematogenic metastases. Over 90% of myxoid liposarcoma are characterized by a reciprocal t(12;16)(q13;p11) translocation. The resulting chimeric FUS–DDIT3 fusion protein plays a crucial role in myxoid liposarcoma pathogenesis; however, its specific impact on oncogenic signaling pathways remains to be substantiated. We here investigate the functional role of FUS–DDIT3 in IGF-IR/PI3K/Akt signaling driving myxoid liposarcoma pathogenesis. Experimental Design: Immunohistochemical evaluation of key effectors of the IGF-IR/PI3K/Akt signaling axis was performed in a comprehensive cohort of myxoid liposarcoma specimens. FUS–DDIT3 dependency and biological function of the IGF-IR/PI3K/Akt signaling cascade were analyzed using a HT1080 fibrosarcoma-based myxoid liposarcoma tumor model and multiple tumor–derived myxoid liposarcoma cell lines. An established myxoid liposarcoma avian chorioallantoic membrane model was used for in vivo confirmation of the preclinical in vitro results. Results: A comprehensive subset of myxoid liposarcoma specimens showed elevated expression and phosphorylation levels of various IGF-IR/PI3K/Akt signaling effectors. In HT1080 fibrosarcoma cells, overexpression of FUS-DDIT3 induced aberrant IGF-IR/PI3K/Akt pathway activity, which was dependent on transcriptional induction of the IGF2 gene. Conversely, RNAi-mediated FUS–DDIT3 knockdown in myxoid liposarcoma cells led to an inactivation of IGF-IR/PI3K/Akt signaling associated with diminished IGF2 mRNA expression. Treatment of myxoid liposarcoma cell lines with several IGF-IR inhibitors resulted in significant growth inhibition in vitro and in vivo. Conclusions: Our preclinical study substantiates the fundamental role of the IGF-IR/PI3K/Akt signaling pathway in myxoid liposarcoma pathogenesis and provides a mechanism-based rationale for molecular- targeted approaches in myxoid liposarcoma cancer therapy. Clin Cancer Res; 23(20); 6227–38. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 12 ( 2019-06-15), p. 3718-3731
    Abstract: Synovial sarcoma is a soft tissue malignancy characterized by a reciprocal t(X;18) translocation. The chimeric SS18-SSX fusion protein acts as a transcriptional dysregulator representing the major driver of the disease; however, the signaling pathways activated by SS18-SSX remain to be elucidated to define innovative therapeutic strategies. Experimental Design: Immunohistochemical evaluation of the Hippo signaling pathway effectors YAP/TAZ was performed in a large cohort of synovial sarcoma tissue specimens. SS18-SSX dependency and biological function of the YAP/TAZ Hippo signaling cascade were analyzed in five synovial sarcoma cell lines and a mesenchymal stem cell model in vitro. YAP/TAZ-TEAD–mediated transcriptional activity was modulated by RNAi-mediated knockdown and the small-molecule inhibitor verteporfin. The effects of verteporfin were finally tested in vivo in synovial sarcoma cell line-based avian chorioallantoic membrane and murine xenograft models as well as a patient-derived xenograft. Results: A significant subset of synovial sarcoma showed nuclear positivity for YAP/TAZ and their transcriptional targets FOXM1 and PLK1. In synovial sarcoma cells, RNAi-mediated knockdown of SS18-SSX led to significant reduction of YAP/TAZ-TEAD transcriptional activity. Conversely, SS18-SSX overexpression in SCP-1 cells induced aberrant YAP/TAZ-dependent signals, mechanistically mediated by an IGF-II/IGF-IR signaling loop leading to dysregulation of the Hippo effectors LATS1 and MOB1. Modulation of YAP/TAZ-TEAD–mediated transcriptional activity by RNAi or verteporfin treatment resulted in significant growth inhibitory effects in vitro and in vivo. Conclusions: Our preclinical study identifies an elementary role of SS18-SSX–driven YAP/TAZ signals, highlights the complex network of oncogenic signaling pathways in synovial sarcoma pathogenesis, and provides evidence for innovative therapeutic approaches.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 388-388
    Abstract: Introduction: Synovial sarcoma (SySa) is a rare soft-tissue malignancy characterized by a specific reciprocal translocation t(X;18). The resulting chimeric SS18-SSX fusion protein acts as transcriptional dysregulator representing the major oncogenic driver in SySa tumorigenesis. Since targeting the fusion protein itself remains a particular challenge, it appears reasonable to therapeutically address signaling pathways which are functionally dependent on the SS18-SSX fusion protein. As different tumor entities were recently shown to harbor aberrant Hippo signaling patterns leading to increased activity of the transcriptional coactivators YAP/TAZ, the aim of this study was to analyze the role of YAP/TAZ in SySa and to decipher the functional link to the SS18-SSX fusion protein. Experimental procedures: YAP/TAZ expression was analyzed by immunohistochemistry in a large cohort of SySa tissue specimens (n=65). Five SySa cell lines and mesenchymal SCP-1 stem cells stably expressing the SS18-SSX fusion protein were employed for in vitro analyses. We set out to analyze whether YAP/TAZ-TEAD transcriptional activity is dependent on the SySa-specific fusion protein, if this dependency is mediated by IGF-IR signaling (known to be activated in SySa) and to understand the biological function of YAP/TAZ in SySa. To modulate YAP/TAZ-TEAD transcriptional activity, RNAi-mediated knockdown and the small molecule inhibitor verteporfin were applied. Finally, the therapeutic effect of YAP/TAZ inhibition was tested in vivo using SySa cell line-based and patient-derived xenografts. Results: SySa tissue specimens and cell lines strongly expressed nuclear YAP/TAZ. RNAi-mediated knockdown of SS18-SSX fusion protein led to significant reduction of YAP/TAZ-TEAD transcriptional activity while SS18-SSX overexpression in SCP-1 cells induced aberrant YAP/TAZ signals. This regulatory connection was at least partly realized through an IGF-II/IGF-IR loop, in which the SS18-SSX fusion protein drives IGF2 expression causing dysregulation of the Hippo effectors LATS1 and MOB1, eventually leading to YAP/TAZ activation. Inhibition of YAP/TAZ-TEAD transcriptional activity by RNAi or verteporfin resulted in a significant induction of apoptosis and significant reduction of SySa cell growth in vitro and in vivo. Conclusions: Our study reveals SS18-SSX fusion protein-driven YAP/TAZ-TEAD signals to play an elementary role in SySa. Given the high efficacy of YAP/TAZ-directed pharmacological approaches in SySa xenografts, this preclinical study may constitute the basis for a novel therapeutic strategy to inhibit SS18-SSX-driven tumorigenesis. Citation Format: Ilka Isfort, Magdalene Cyra, Sandra Elges, Sareetha Kailayangiri, Bianca Altvater, Claudia Rossig, Jan-Henrik Mikesch, Agnieszka Wozniak, Patrick Schöffski, Eva Wardelmann, Marcel Trautmann, Wolfgang Hartmann. SS18-SSX modulates YAP/TAZ-TEAD transcriptional activity in synovial sarcoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 388.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages