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: Life Science Alliance, Life Science Alliance, LLC, Vol. 1, No. 5 ( 2018-10), p. e201800190-
    Abstract: The contribution of the tumor microenvironment to pancreatic ductal adenocarcinoma (PDAC) development is currently unclear. We therefore examined the consequences of disrupting paracrine Hedgehog (HH) signaling in PDAC stroma. Herein, we show that ablation of the key HH signaling gene Smoothened ( Smo ) in stromal fibroblasts led to increased proliferation of pancreatic tumor cells. Furthermore, Smo deletion resulted in proteasomal degradation of the tumor suppressor PTEN and activation of oncogenic protein kinase B (AKT) in fibroblasts. An unbiased proteomic screen identified RNF5 as a novel E3 ubiquitin ligase responsible for degradation of phosphatase and tensin homolog (PTEN) in Smo -null fibroblasts. Ring Finger Protein 5 ( Rnf5 ) knockdown or pharmacological inhibition of glycogen synthase kinase 3β (GSKβ), the kinase that marks PTEN for ubiquitination, rescued PTEN levels and reversed the oncogenic phenotype, identifying a new node of PTEN regulation. In PDAC patients, low stromal PTEN correlated with reduced overall survival. Mechanistically, PTEN loss decreased hydraulic permeability of the extracellular matrix, which was reversed by hyaluronidase treatment. These results define non-cell autonomous tumor-promoting mechanisms activated by disruption of the HH/PTEN axis and identifies new targets for restoring stromal tumor-suppressive functions.
    Type of Medium: Online Resource
    ISSN: 2575-1077
    Language: English
    Publisher: Life Science Alliance, LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2948687-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Neoplasia, Elsevier BV, Vol. 18, No. 9 ( 2016-09), p. 541-552
    Type of Medium: Online Resource
    ISSN: 1476-5586
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2016
    detail.hit.zdb_id: 2008231-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2883-2883
    Abstract: Purpose of Study Pancreatic cancer is an overwhelming fatal disease with less than 5% of patients surviving beyond 5 years. The most prominent histopathological hallmark of pancreatic cancer is its uniquely dense stromal reaction as evidenced by recent reports that highlight the significant role of stromal fibroblasts on pancreatic tumor cell biology. We used novel mouse models to show that genetic inactivation of Smoothened (Smo) in stromal fibroblasts accelerated Kras-initiated tumorigenesis. Research Method We used a genetically engineered mouse model of pancreatic cancer that relies on constitutive activation of the Kras oncogene in the epithelium. We simultaneously employed cre-loxP technology to conditionally delete Smo exclusively in the fibroblast compartment of the pancreas, thus disrupting the crucial hedgehog paracrine signaling loop between pancreatic tumor cells and fibroblasts. Novel Findings We showed that deletion of Smo in stromal fibroblasts accelerated pancreatic tumorigenesis through a mechanism involving destabilization of fibroblast PTEN protein. An unbiased genetic screen revealed the ubiquitin E2 conjugating enzyme UBE2K as a PTEN destabilizer and knockdown of UBE2K blocked the degradation of PTEN protein. Down-regulation of PTEN enhanced TGF-α production in stromal fibroblasts, and increased epithelial cell transformation and proliferation through epithelial growth factor receptor (EGFR). A selective SMO inhibitor also decreased PTEN in a Kras mouse model as well as in human primary pancreatic cancer associated fibroblasts. Importantly, in pancreatic ductal adenocarcinoma (PDAC) patient samples, low PTEN expression correlated with low SMO expression and with reduced overall survival. These results define a pathway that reprograms stromal fibroblasts from a tumor suppressive phenotype to a tumor promoting phenotype, thus highlighting the dual functions of stromal fibroblasts in pancreatic cancer and the molecular consequences of loss of the hedgehog pathway. Thus, a more comprehensive understanding of tumor-stroma interactions is required to assure effective implementation of targeted therapies. Conclusions & Implications Recent pre-clinical reports suggest the pancreatic tumor microenvironment functions predominantly to inhibit tumor growth, challenging the concept of tumor stroma as a therapeutic target. Our results provide molecular insight into how the balance between the opposing activities of tumor stromal fibroblasts is maintained, and potentially identifies targets for restoring stromal tumor suppressive functions. In summary, we demonstrate that ablation of paracrine hedgehog signaling in SMA-positive fibroblasts leads to proteasome-mediated degradation of the PTEN tumor suppressor protein and subsequent activation of oncogenic pathways. Citation Format: Jason R. Pitarresi, Jinghai Wu, Xin Liu, Veronica Bravo, Maria C. Cuitiño, Raleigh D. Kladney, Sarah Woelke, Sarmila Majumder, Gustavo Leone, Michael C. Ostrowski. Genetic ablation of Smoothened in tumor-associated fibroblasts promotes pancreatic tumorigenesis. [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 2883. doi:10.1158/1538-7445.AM2015-2883
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    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 ...
  • 4
    In: Genes & Development, Cold Spring Harbor Laboratory, Vol. 30, No. 17 ( 2016-09-01), p. 1943-1955
    Abstract: The contribution of the microenvironment to pancreatic acinar-to-ductal metaplasia (ADM), a preneoplastic transition in oncogenic Kras -driven pancreatic cancer progression, is currently unclear. Here we show that disruption of paracrine Hedgehog signaling via genetic ablation of Smoothened ( Smo ) in stromal fibroblasts in a Kras G12D mouse model increased ADM. Smo -deleted fibroblasts had higher expression of transforming growth factor-α ( Tgfa ) mRNA and secreted higher levels of TGFα, leading to activation of EGFR signaling in acinar cells and increased ADM. The mechanism involved activation of AKT and noncanonical activation of the GLI family transcription factor GLI2. GLI2 was phosphorylated at Ser230 in an AKT-dependent fashion and directly regulated Tgfa expression in fibroblasts lacking Smo . Additionally, Smo -deleted fibroblasts stimulated the growth of Kras G12D / Tp53 R172H pancreatic tumor cells in vivo and in vitro. These results define a non-cell-autonomous mechanism modulating Kras G12D -driven ADM that is balanced by cross-talk between Hedgehog/SMO and AKT/GLI2 pathways in stromal fibroblasts.
    Type of Medium: Online Resource
    ISSN: 0890-9369 , 1549-5477
    RVK:
    Language: English
    Publisher: Cold Spring Harbor Laboratory
    Publication Date: 2016
    detail.hit.zdb_id: 1467414-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: PLOS ONE, Public Library of Science (PLoS), Vol. 12, No. 9 ( 2017-9-21), p. e0184984-
    Type of Medium: Online Resource
    ISSN: 1932-6203
    Language: English
    Publisher: Public Library of Science (PLoS)
    Publication Date: 2017
    detail.hit.zdb_id: 2267670-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 13_Supplement ( 2015-07-01), p. B01-B01
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 13_Supplement ( 2015-07-01), p. B01-B01
    Abstract: Purpose of Study:Pancreatic cancer remains an overwhelmingly fatal disease with approximately 95% of patients dying within 5 years of diagnosis. Many recent reports have highlighted the emerging role that stromal cells’fibroblasts in particular’have on pancreatic tumor cell biology. It has become apparent that new models of pancreatic cancer that accurately portray the tumor microenvironment (TME) are necessary to move the field forward. Advances in genetic tools to modulate the TME developed in our lab have allowed us the unique opportunity to manipulate specific stromal cell compartments. Our overarching hypothesis is: Ets-2 in the pancreatic tumor-associated stroma promotes tumor initiation and development by regulating essential signaling pathways in fibroblasts and is critical for tumor-stroma co-evolution. Research Method: In this study we use a previously validated and well-studied genetically engineered mouse model of pancreatic cancer that relies on constitutive activation of the Kras oncogene tumor cell lineage. We simultaneously employ lox-cre technology to conditionally delete our gene of interest exclusively in the fibroblast compartment of the pancreas. For this study we delete the transcription factor Ets-2, which has previously been shown to effect tumor initiation and growth in mammary stroma. Novel Findings: Here we show that Ets-2 in pancreatic tumor-associated stroma promotes tumor formation by regulating essential signaling pathways in stromal fibroblasts and is critical for tumor-stroma co-evolution. We conditionally deleted Ets-2 in cancer associated fibroblasts (CAFs), thus altering stroma-tumor crosstalk and resulting in delayed tumor initiation. Specifically, we saw a decrease in pre-cancerous acinar-to-ductal metaplasic (ADM) and pancreatic intraepithelial neoplastic (PanIN) lesions in mice null for Ets-2 in the fibroblast compartment. In order to investigate the mechanism by which Ets-2 is able to effect progression of tumors from the fibroblast compartment, we developed a method for harvesting and purifying primary pancreatic CAFs. We performed microarray and gene expression analysis on Ets-2 deleted CAFs and saw significantly decreased expression of secreted factors. The altered secretome upon Ets-2 deletion in fibroblasts was crucially lacking tumor necrosis factor alpha (TNFα), a known pro-tumor ligand in pancreatic carcinogenesis. Furthermore, we found evolutionarily conserved Ets-2 transcription factor binding sites in the proximal promoter of TNFα, suggesting that Ets-2 directly regulates TNFα transcription. Thus we have shown that Ets-2 ablation in pancreatic fibroblasts delays pancreatic tumor initiation through the pro-tumor ligand TNFα. Conclusions and Implications:This report shows that deleting a gene in pancreatic fibroblasts causes a change in tumor-stroma co-evolution and that Ets-2 is able to act as a novel oncogene in cancer associated fibroblasts to promote pancreatic carcinogenesis. We have shown that Ets-2 deletion significantly changed the biological role of the CAFs and negatively effects tumor growth. This finding is relevant to the field of pancreatic cancer because it shows that the pancreatic TME can play a driver role in overall tumor initiation and development. These collective findings contribute to our lab’s overall hypothesis that the tumor microenvironment is not a mere bystander or byproduct of tumor development, but rather that it can drive tumor evolution amongst a variety of cancers. Citation Format: Jason R. Pitarresi, Jinghai Wu, Sarah Woelke, Raleigh Kladney, Maria Cuitino, Lianbo Yu, Ostrowski C. Michael. Ets-2 acts as a novel oncogene in cancer associated fibroblasts and promotes pancreatic tumor initiation and development. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Innovations in Research and Treatment; May 18-21, 2014; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2015;75(13 Suppl):Abstract nr B01.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    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...