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
Filter
  • American Association for Cancer Research (AACR)  (6)
  • Nakai, Yousuke  (6)
Material
Publisher
  • American Association for Cancer Research (AACR)  (6)
Language
Years
Subjects(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 24_Supplement ( 2016-12-15), p. B66-B66
    Abstract: Background and Aims: Pancreatic ductal adenocarcinoma (PDAC) is well characterized by dense fibrotic stroma with abundant cancer-associated fibroblasts (CAFs). As CAFs are activated during tumorigenesis and acquire tumor-promoting properties, activated CAFs have been implicated in PDAC progression; however, the precise mechanisms of their activation remain largely unknown. The bromodomain and extraterminal (BET) domain proteins are epigenetic reader proteins that recognize acetylated amino acid residues on histone tails and facilitate gene transcription. Recent studies have demonstrated therapeutic efficacy of BET inhibitors on various cancers including PDAC, mainly through suppression of c-myc transcription; however, how BET inhibitors suppress PDAC growth and their effects on CAFs remains largely unknown. Using patient-derived tumor xenografts (PDX) and primary CAFs, we investigated the therapeutic efficacy and dissected the underlying mechanisms of a BET inhibitor, JQ1, on human PDAC and CAFs. Methods: We established PDX lines and primary CAFs from surgically resected human PDAC specimen. For in vivo analyses, mice bearing subcutaneous tumor were treated with vehicle or JQ1. For in vitro analyses, patient-derived PDAC cells and CAFs were treated with vehicle or JQ1 and analyzed separately. To explore the pro-tumorigenic role of secretion from CAFs, PDAC cells were cultured with conditioned medium (CM) that was collected from DMSO- or JQ1- treated CAFs. Chromatin immunoprecipitation (ChIP) assay was performed to assess the binding of transcription factors and histone modifications which are associated with altered gene expression in CAFs by JQ1 treatment. Results: In vivo experiments revealed that volumes and weights of subcutaneous PDX tumors were significantly smaller in JQ1-treated mice than vehicle-treated mice. Unexpectedly, however, JQ1 exerted only minimal effects to the proliferation of PDAC cells that were isolated from PDX tumors and cultured in vitro, suggesting the involvement of cell-extrinsic mechanisms in the JQ1-mediated suppression of tumor growth in vivo. Of note, histopathological analysis of PDX tumors revealed that JQ1 treatment dramatically ameliorated desmoplastic change, with reduction in extracellular matrix (ECM) deposition and α-SMA expressing CAFs. As α-SMA expression and ECM production is a hallmark of activated CAFs, we hypothesized that JQ1 might inactivate CAFs, thereby reducing their tumor-promoting properties. To test this hypothesis, qPCR was performed to analyze gene expression in primary CAFs cultured in vitro and also in stromal cells in PDX tumors in vivo. As expectedly, JQ1 suppressed the expression of genes implicated in the properties of activated CAF, including ECM, cytokines and growth factors both in vitro and in vivo. Furthermore, when PDAC cells were cultured with CM from DMSO–treated CAFs, proliferation of PDAC cells were promoted along with activation of MAPK, AKT, and STAT3 pathways, which was abrogated when cultured with CM from JQ1-treated CAFs. Consistently, immunoblotting and immunohistochemistry of PDX tumors demonstrated that JQ1 reduced phosphorylation of ERK, AKT, and STAT3 in PDAC cells in vivo. Mechanistically, we found that JQ1 suppressed hedgehog and TGF-β/SMAD3 pathways, both of which play central roles in CAF activation, through disruption of BRD4 recruitment to the promoter regions of their target genes. Conclusions: BET proteins are critical regulators of CAF-activation in PDAC. Inactivation of CAFs by BET inhibition offers a novel therapeutic approach for PDAC. Citation Format: Keisuke Yamamoto, Keisuke Tateishi, Yotaro Kudo, Mayumi Hoshikawa, Mariko Tanaka, Takuma Nakatsuka, Hiroaki Fujiwara, Koji Miyabayashi, Ryota Takahashi, Yasuo Tanaka, Hideaki Ijichi, Yousuke Nakai, Hiroyuki Isayama, Yasuyuki Morishita, Taku Aoki, Yoshihiro Sakamoto, Kiyoshi Hasegawa, Norihiro Kokudo, Masashi Fukayama, Kazuhiko Koike.{Authors}. BET inhibition remodels tumor stroma and suppresses progression of human pancreatic cancer. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2016 May 12-15; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2016;76(24 Suppl):Abstract nr B66.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 7 ( 2013-04-01), p. 2221-2234
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly cancers worldwide. Although many regimens have been used for PDAC treatment, the combination of the EGF receptor (EGFR) inhibitor erlotinib with gemcitabine has been the only molecular-targeted drug tested so far that has been superior to gemcitabine alone. The mechanism underlying this effective combinational regimen remains unknown. Here, we show that the combination is superior to gemcitabine alone in blocking progression and prolonging survival in a murine model of PDAC (Kras activation with Tgfbr2 knockout). We found that gemcitabine induced mitogen-activated protein kinase signaling, which was dramatically inhibited by erlotinib even in the Kras-activated PDAC cells in the mouse model. Mechanistic investigations suggested that gemcitabine induces EGFR ligand expression and ERBB2 activation by increasing heterodimer formation with EGFR, thereby maintaining high levels of ERBB2 protein in PDAC cells. Overall, our findings suggest a significant role of ERBB in PDAC treatment. Cancer Res; 73(7); 2221–34. ©2013 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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 ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 851-851
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly cancers. New therapeutic approaches to improve the prognosis are highly required. We generated pancreas-specific TGF-beta receptor II (Tgfbr2) knockout mice in the context of Kras activation, which recapitulated clinical and histopathological manifestations of human PDAC, indicating that an impact of blockade of TGF-beta signaling in PDAC progression. An EGFR inhibitor erlotinib showed a superior survival effect in combination with gemictabine, however, the mechanism by which PDAC with extremely frequent KRAS-mutation benefits from EGFR inhibition remains largely unkown. Using the model we investigated the anti-tumor impact and mechanisms of erlotinib in combination with gemcitabine. We treated the mice with elrotinib and gemcitabine. Median survival times were 52.5, 69, and 74 days for control, gemcitabine alone, and gemcitabine + erlotinib, respectively. Gemcitabine alone significantly extended the survival compared to the control. Further, adding erlotinib to gemcitabine prolonged the survival significantly compared to gemcitabine alone. Interestingly, treatment with gemcitabine enhanced the activation of MAPK signaling, which was inhibited by adding erlotinib, even in the KRAS-mutant PDAC in vivo and in vitro. We observed that the gemcitabine-induced MAPK signaling activation was due to induction of EGFR ligands expression and also due to ERBB2 activation in PDAC cells. Erlotinib inhibited the ERBB2 activation, partly by inhibiting heterodimer formation with EGFR and also decreasing ERBB2 protein expression in PDAC cells. This mouse model appears recapitulating chemosensitivity of human PDAC and indicates the significance of ERBBs in PDAC treatment. This model also might provide important insights into the beneficial patient subpopulation and the mechanism of the action of drugs and chemoresistance, in which TGF-beta signaling might be highly involved. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 851. doi:1538-7445.AM2012-851
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 2_Supplement ( 2016-01-15), p. A40-A40
    Abstract: The role of genetic mutations in the pathogenesis of pancreatic ductal adenocarcinoma (PDAC) is well established. However, it is still unclear if epigenetic aberrations contribute to PDAC progression. We previously reported a novel role for the H3K27 demethylase KDM6B/JMJD3 in regulating PDAC progression (Carcinogenesis 2014;35(11):2404-14). KDM6B was downregulated in high grade PDACs and knockdown (KD) of KDM6B in PDAC cells increased the tumorigenicity and enhanced the aggressive phenotypes of these cells in vivo. Furthermore, CCAAT enhancer binding protein alpha gene (CEBPA) was identified as a direct target of KDM6B, and reduced KDM6B- C/EBPα axis was resulted in increased aggressiveness in PDAC cells. To dissect further the pathological effects caused by loss of KDM6B- C/EBPα function in PDAC cells, we tried to identify a surrogate molecular marker of the cells lacking of KDM6B- function. For the purpose, we used a cDNA microarray to compare the expression profiles of KDM6B- KD and control BxPC3 PDAC cells. 906 genes were upregulated and 639 downregulated in KDM6B- KD BxPC3 cells compared to the control cells. We focused on 58 genes encoding cell-surface molecules that were upregulated in KDM6B- KD BxPC3 cells and validated the expression of 9 surface marker candidates, including 3 that have already been reported to be expressed on PDAC tumor-initiating cells, namely, CD24, CD44, and CD133. Only CD47 was significantly upregulated in the KDM6B- KD BxPC3 cells as confirmed by both quantitative RT-PCR and flow cytometric analysis. CD47 was also upregulated in other PDAC cell lines following KDM6B knockdown. It has recently been reported that CD47 is upregulated in various malignancies and that an increase in CD47 expression is correlated with a poor prognosis. In line with the previous reports, CD47high cells formed about 4-fold more spheres than non-CD47high cells. The close relationship between CD47 expression and the sphere-forming ability was supported by the finding that CD47low cells formed even fewer spheres. To confirm these results in vivo, the sorted CD47high and non-CD47high cells were subcutaneously xenotransplanted into nude mice. All CD47high cells formed tumors more efficiently than the unfractionated KDM6B- KD cells, while the tumor-forming rate of non-CD47high cells was comparable to that of the Ctrl cells. In addition, when the cells were injected into the spleens of nude mice, CD47high cells demonstrated higher liver metastatic potential than the non-CD47high population. These data suggested that the increased tumor-initiating potential of KDM6B- KD cells was attributable to this induced CD47high population. Consistently, the expression of KDM6B and C/EBPα inversely correlated with CD47 expression and tumor grade in human PDAC tumors. Collectively, our data provides a link between epigenetic change and PDAC progression, thus offering a novel strategy to target PDAC aggressiveness by intervening in the dynamics of epigenetic process. Citation Format: Keisuke Yamamoto, Keisuke Tateishi, Yotaro Kudo, Koji Miyabayashi, Ryota Takahashi, Takuma Nakatsuka, Hiroaki Fujiwara, Yousuke Nakai, Yasuo Tanaka, Hideaki Ijichi, Hiroyuki Isayama, Kazuhiko Koike. Emergence of CD47- high expression cells confers enhanced tumorigenicity upon KDM6B suppression in pancreatic cancer. [abstract]. In: Proceedings of the AACR Special Conference on Chromatin and Epigenetics in Cancer; Sep 24-27, 2015; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2016;76(2 Suppl):Abstract nr A40.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 13_Supplement ( 2015-07-01), p. A55-A55
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 13_Supplement ( 2015-07-01), p. A55-A55
    Abstract: TGF-beta signaling has a crucial role in pancreatic tumorigenesis, and almost all of pancreatic cancers carry at least one genetic alteration of TGF-beta related genes, such as SMAD4, TGFBR2, SMAD3, and BMPR2. However, the role of BMP signaling in pancreatic cancer remains unclear. Previous studies reported the depletion of BMP signaling resulted in the aggressive phenotype of cancer, and some reported BMP signaling played an important role in tumor progression and metastasis. We have already established pancreas-specific Tgbr2 knockout mice in the context of Kras activation, which clinically and histopathologically recapitulate human PDAC. With regard to PDAC, Smad4 mutation or deletion is more commonly observed, however the Smad4 knockout mice with activating Kras mutation was reported to show cystic type tumor of pancreas. Therefore, our Kras+Tgfbr2KO might be the closest approximation of the human PDAC in terms of histology. We examined the effect of BMP signaling on the tumorigenesis and progression of PDAC using this mouse model. We performed immunohistochemistry of murine PDAC to evaluate whether BMP signaling was related to the PDAC progression. We examined the effect of Bmp4 and Bmp7 on the proliferation, invasion and adhesion using murine PDAC and PanIN cells in vitro. We have already established the murine PDAC cell lines from Pancreas-specific Kras+Tgfbr2KO mice and murine PanIN cells from Pancreas-specific activating Kras mutation mice. Bmpr2 was knocked down in PanIN cell lines using shRNA, and we examined whether the effect of BMP signaling was canceled by Bmpr2 knockdown. In vivo, we evaluated the effect of BMP signaling on tumor growth and tumor-stromal interaction using the xenograft mouse model of Bmpr2-negative PanIN cells. The immunohistochemistry of murine pancreas tissues demonstrated that Smad1/5/8 was more strongly phosphorylated in PDAC compared to PanIN lesion. We also observed that Smad1/5/8 was phosphorylated in stromal cells surrounding tumor areas, which was likely to suggest the importance of BMP signaling in PDAC progression and tumor-stromal interaction. In vitro, both Bmp4 and Bmp7 did not affect the proliferation and invasion of PDAC and PanIN cells, but they increased the adhesion of PDAC and PanIN cells, and knockdown of Bmpr2 canceled the effect of Bmps. In vivo, we evaluated the growth of subcutaneous tumor allograft and the tumors of Bmpr2-negative PanIN cells showed slower tumor growth than tumors of the control, differently from the results in vitro. These results suggested that BMP signaling was associated with the tumor-stromal interaction and played important role in tumor progression. In this study we evaluated the role of BMP signaling in pancreatic cancer using pancreas-specific Kras+Tgfbr2KO mice, and demonstrated that BMP signaling played important role in the adhesion and progression of pancreatic cancer, which was due to the tumor-stromal interaction. Citation Format: Koji Miyabayashi, Hideaki Ijichi, Ryota Takahashi, Keisuke Yamamoto, Yoshinari Asaoka, Keisuke Tateishi, Yousuke Nakai, Hiroyuki Isayama, Harold L. Moses, Kazuhiko Koike. A role of bone morphogenetic protein signaling in pancreatic cancer. [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 A55.
    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 ...
  • 6
    In: Cancer Epidemiology, Biomarkers & Prevention, American Association for Cancer Research (AACR), Vol. 30, No. 5 ( 2021-05-01), p. 1020-1028
    Abstract: ABO blood group has been associated with risks of various malignancies, including pancreatic cancer. No study has evaluated the association of ABO blood group with incidence of pancreatic carcinogenesis during follow-up of patients with intraductal papillary mucinous neoplasms (IPMN). Methods: Among 3,164 patients diagnosed with pancreatic cysts at the University of Tokyo (Tokyo, Japan) from 1994 through 2019, we identified 1,815 patients with IPMN with available data on ABO blood group. We studied the association of ABO blood group with incidence of pancreatic carcinoma, overall and by carcinoma types [IPMN-derived carcinoma or concomitant pancreatic ductal adenocarcinoma (PDAC)]. Utilizing competing-risks proportional hazards models, we estimated subdistribution hazard ratios (SHR) for incidence of pancreatic carcinoma with adjustment for potential confounders, including cyst characteristics. Results: During 11,518 person-years of follow-up, we identified 97 patients diagnosed with pancreatic carcinoma (53 with IPMN-derived carcinoma and 44 with concomitant PDAC). Compared with patients with blood group O, patients with blood groups A, B, and AB had multivariable SHRs (95% confidence intervals) for pancreatic carcinoma of 2.25 (1.25–4.07; P = 0.007), 2.09 (1.08–4.05; P = 0.028), and 1.17 (0.43–3.19; P = 0.76), respectively. We observed no differential association of ABO blood group with pancreatic carcinoma incidence by carcinoma types. Conclusions: In this large long-term study, patients with IPMN with blood group A or B appeared to be at higher risk of pancreatic carcinoma compared with those with blood group O. Impact: ABO blood group can be a biomarker for pancreatic cancer risk among patients with IPMNs.
    Type of Medium: Online Resource
    ISSN: 1055-9965 , 1538-7755
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036781-8
    detail.hit.zdb_id: 1153420-5
    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...