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)  (13)
  • 2015-2019  (13)
  • Medicine  (13)
Material
Publisher
  • American Association for Cancer Research (AACR)  (13)
Language
Years
  • 2015-2019  (13)
Year
Subjects(RVK)
  • Medicine  (13)
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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 4002-4002
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 4002-4002
    Abstract: Introduction: While the process of carcinogenesis is generally a multistep with accumulation of mutations in oncogenes and tumor-suppressor genes, pancreatic cancers have frequent mutations relatively in limited genes, such as KRAS, TP53 (p53), CDKN2A (p16), and SMAD4. Then, series of mouse models have been generated to target these genes in the pancreas, and pancreatic cancers or pre-cancerous tumors have been successfully observed in such gene-modified mice. These mouse models are advantageous compared to human samples in genetic simplicity and little influence of environmental factors. During the steps of carcinogenesis, microRNAs (miRNAs), regulators of gene expression, may also be dysregulated. Although many studies have already been performed to determine the dysregulated miRNAs in pancreatic cancers, consistent results have not always been obtained so far, probably due to the complexity of the samples tested. Then, in this study, we examined comprehensive expression profiles of miRNAs in the pancreatic tissues in two kinds of gene-modified mice, which develop pre-cancerous tumor or progressive pancreatic cancer, to obtain an information about the expression changes of miRNA levels during the pancreatic multistep carcinogenesis. Methods: We used two mouse models: Ptf1a cre/+; LSL-Kras G12D/+ which induced constitutively active Kras mutation specifically in the pancreas and develop focal premalignant ductal tumors similar to human pancreatic intraepithelial neoplasia (PanINs). Ptf1acre/+;LSL-KrasG12D/+;Tgfbr2flox/flox, which induced Tgfbr2 knockout with active Kras expression in the pancreas and promoted aggressive pancreatic carcinoma with similar histology to human pancreatic ductal adenocarcinoma (PDAC). We performed miRNA microarrays (3D-GENE mouse miRNA Oligo chip, Toray) using total RNAs from pancreatic tumors of these mouse models and from wild type pancreas tissues. Results; MiRNAs, such as miR-21, miR-125b-5p, miR-31, and miR-192, were upregulated in PDAC compared to normal tissues. Interestingly, the expression levels of the majority of these miRNAs already increased from the stage of pre-cancerous PanINs in Kras-mutated mice. However, some miRNAs, such as miR-669p* and miR-200b/c, were increased at the stage between PDAC and PanINs, while no changes were observed at the stage of PanINs. In contrast, miR-148a and miR-802 were downregulated from the stage of PanINs, and miR-187* decreased during the stages of between PDAC and PanINs. Discussion; We showed using genetically-simple mouse models that the changes of miRNA expression levels occur at specific stages during the course of pancreatic carcinogenesis. Although validation of these results is required in human samples, the stage-specific expression profiles of miRNAs may provide with the insights regarding the pathogenesis of pancreatic multistep-carcinogenesis, and the information may also be useful as novel diagnostic biomarkers. Citation Format: Takahiro Kishikawa, Motoyuki Otsuka, Takeshi Yoshikawa, Motoko Ohno, Kazuhiko Koike. Alternations in microRNA expression profiles during the pancreatic multistep carcinogenesis. [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 4002. doi:10.1158/1538-7445.AM2015-4002
    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 ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 20 ( 2019-10-15), p. 6217-6227
    Abstract: Hepatitis B viral (HBV) DNA is frequently integrated into the genomes of hepatocellular carcinoma (HCC) in patients with chronic HBV infection (chronic HBV, hereafter), whereas the frequency of HBV integration in patients after the disappearance of HBV (prior HBV, hereafter) has yet to be determined. This study aimed to detect integration of HBV and adeno-associated virus type 2 (AAV2) into the human genome as a possible oncogenic event. Experimental Design: Virome capture sequencing was performed, using HCC and liver samples obtained from 243 patients, including 73 with prior HBV without hepatitis C viral (HCV) infection and 81 with chronic HBV. Results: Clonal HBV integration events were identified in 11 (15.0%) cases of prior HBV without HCV and 61 (75.3%) cases of chronic HBV (P & lt; 0.001). Several driver genes were commonly targeted by HBV, leading to transcriptional activation of these genes; TERT [four (5.4%) vs. 15 (18.5%)], KMT2B [two (2.7%) vs. five (6.1%)] , CCNE1 [zero vs. one (1.2%)], CCNA2 [zero vs. one (1.2%)] . Conversely, CCNE1 and CCNA2 were, respectively, targeted by AAV2 only in prior HBV. In liver samples, HBV genome recurrently integrated into fibrosis-related genes FN1, HS6ST3, KNG1, and ROCK1 in chronic HBV. There was not history of alcohol abuse and 3 patients with a history of nucleoside analogue treatment for HBV in 8 prior HBV with driver gene integration. Conclusions: Despite the seroclearance of hepatitis B surface antigen, HBV or AAV2 integration in prior HBV was not rare; therefore, such patients are at risk of developing HCC.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 730-730
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 730-730
    Abstract: Pancreatic ductal adenocarcinoma (Pdac) is one of the most intractable malignancies due to difficulties in early detection. Although promising biomarkers are increasingly reported, such methods are not yet easy to apply clinically, mainly due to their low reproducibility or technical difficulties. In this study, we developed a convenient and sensitive method for quantifying aberrantly expressed satellite repeat RNAs in sera, which can be used to efficiently detect patients with Pdac. It is difficult to correctly quantitate repeat arrays because its repetitive nature makes it difficult to establish appropriate primers that amplify a single product using simple PCR procedures. Here, we introduce a Tandem Repeat Amplification by nuclease Protection (TRAP) method combined with droplet digital PCR (ddPCR) to detect human satellite II (HSATII) RNAs, which are specifically expressed in human Pdacs at greater levels than normal tissues. HSATII RNA core sequence levels in sera were significantly higher in Pdac patients compared with non-cancer patients (median copy number: 14.75 and 3.17 per µl in the training set and 17.35 and 2.9 in the validation set, respectively). In addition, patients with intraductal papillary mucinous neoplasm (IPMN), a precancerous lesion of Pdac, could also be efficiently detected. This method can be routinely applied to screen patients with Pdac and high-risk patients, facilitating the development of preventive medicine for this disease. Citation Format: Takahiro Kishikawa, Motoyuki Otsuka, Kazuhiko Koike. High sensitive detecting procedure of circulating repetitive RNA as novel early marker of pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 730. doi:10.1158/1538-7445.AM2017-730
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 3339-3339
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 3339-3339
    Abstract: Within the gastrointestine, nerves help to regulate both normal and neoplastic stem cell dynamics. Several previous studies suggested that cholinergic nerve signaling plays an important role in gastrointestinal cancer development, but the exact underlying mechanism has not been clarified. In this study, we examined the role of muscarinic acetylcholine receptor subtype 3 (M3R) in gastric homeostasis and cancer development by using mouse models and human cancer cell lines. In situ hybridization revealed M3R expression in gastric stem cell zone, and its expression was markedly upregulated in gastric cancer cells. We knocked out M3R in Lgr5+ gastric stem cells in Lgr5-CreERT; M3Rflox/flox mice, and found that deletion of M3R inhibited clonal expansion of Lgr5+ cells in regenerative states. In a gastric tumor model of Mist1-CreERT; Apcflox/flox mice, knockout of M3R dramatically suppressed tumor growth. RNA sequencing analysis of these tumors revealed that several important pathways were significantly inhibited in M3R knockout samples, including YAP/TAZ pathway. We established M3R-expressing gastric cancer cell lines, and western blotting, luciferase assay, and RT-PCR analysis confirmed that acetylcholine (ACh) agonist activates YAP pathway through M3R. YAP is upregulated in approximately half of gastric cancer patients, and its expression is significantly associated with disease stage and histological form. This M3R-YAP axis activates the gastric stem cell niche and offers a compelling target for tumor treatment and prevention. Note: This abstract was not presented at the meeting. Citation Format: Yoku Hayakawa, Mitsuru Konishi, Kosuke Sakitani, Kazuhiko Koike, Timothy Wang. Muscarinic acetylcholine receptor subtype 3 regulates gastric stem cell expansion and gastric cancer progression by controlling YAP activation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3339. doi:10.1158/1538-7445.AM2017-3339
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 912-912
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 912-912
    Abstract: Intestinal crypts are maintained by long-lived intestinal stem cells (ISCs) which reside near the base of glands. Above the ISC pool, there are short-lived progenitors that can supply lineage-specific differentiated cell types into the villus. Notch and Wnt pathways play a key role for determining the stem/progenitor cell function and their cell fate, and regulating cancer development in the gastrointestine. Although it has been well established that ISCs are a major origin of intestinal cancer, it remains unknown whether intestinal progenitor population can give rise to cancer. We use Mist1-CreERT mice and induce Notch and Wnt activation by mating with LSL-Notch1-IC mice and/or Apc flox mice. We found that a bHLH transcriptional factor Mist1 is expressed in Paneth cells (Lysozyme+CD24high) as well as short-lived secretory progenitors (Lysozyme-CD24low) in the intestine. Mist1+ secretory progenitors are radio- and chemo-resistant, but do not show stem cell interconversion even after intestinal injury or Lgr5 ablation. However, aberrant Notch activation changes Mist1+ cells to Lgr5+ long-lived enterocyte progenitors, with loss of secretory lineage differentiation. Mist1+CD24low progenitor population can form intestinal organoids with Notch activation in vitro. Mist1+ secretory progenitors can give rise to intestinal cancer by simultaneous Notch and Wnt activation by loss of Apc gene, while loss of Apc in Mist1 lineage alone does not develop cancer. In the colon, Mist1 marks colonic secterory progenitors that become to lineage trace and can be a cancer-initiating cell after Notch activation or DSS-induced colonic injury. These results provide the clear evidence of cellular plasticity dependent on Notch signaling in the gut, and suggest short-lived progenitors as another cellular origin of cancer besides ISC pool. Citation Format: Yoku Hayakawa, Kosuke Sakitani, Woosook Kim, Yagnesh Tailor, Karan Nagar, Kazuhiko Koike, Samuel Asfaha, Timothy C. Wang. Mist1+ secretory progenitor cells can give rise to cancer in the intestine and colon. [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 912.
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 962-962
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 962-962
    Abstract: Highly repetitive tandem arrays located at centromere and pericentromere regions of the chromosomes had been considered epigenetically silent by heterochromatin modification. However, the deregulated transcription of the non-coding satellite sequences exists in human and mouse pancreatic cancer tissues. This aberrant expression can be detected even in Kras-mutated pancreatic intraepithelial neoplasia (PanIN) tissues, which are pre-cancerous lesions to invasive pancreatic cancer. To examine the biological roles of these aberrantly expressed satellite RNAs during the carcinogenesis steps, we established genetically-modified mouse PanIN-derived cells, in which mouse major satellite (MajSAT) RNAs were ectopically expressed. MajSAT RNA expressing PanIN-derived cells showed the increased chromosomal instability and the rate of spontaneous point mutations in the genomic as well as in the mitochondrial DNAs. We identified YBX1 protein as a binding protein specifically with MajSAT RNA by RNA immunoprecipitation. MajSAT RNA inhibited nuclear translocation of YBX1 under oxidative damage, which reduced the DNA damage repair activity of YBX1. The mutation rate of genomic and mitochondrial DNAs, and transformation rate were rescued by YBX1 overexpression in these cells. These findings indicate that satellite transcripts at the early stage of cancer development may act as “mutagens” and accelerate oncogenic processes. Citation Format: Takahiro Kishikawa, Motoyuki Otsuka, Kazuhiko Koike. Deregulated transcription of mouse satellite sequences accelerates oncogenic processes via functional inhibition of YBX1 protein. [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 962.
    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 ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 4456-4456
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 4456-4456
    Abstract: Introduction: Hepatocellular carcinoma (HCC) is an important issue to public health of the world. By a genome-wide association study, we previously identified that a single nucleotide polymorphism (SNP) in the promoter region of the MHC class I polypeptide-related sequence A (MICA) gene is significantly associated with the risk of hepatitis-virus-related HCC. MICA is a ligand for NKG2D, a cell receptor of NK cells, which activates NK cells to clear the pathogenic cells expressing MICA such as infected cells or malignant cells as a “danger” signal. However, unexpectedly, the Hepatitis B virus (HBV)-infected patients with risk allele of MICA gene showed higher levels of serum MICA. This suggested that the serum MICA, which is excreted from the infected hepatocytes by a shedding mechanism, may work as a decoy for NK cells and inhibit their activities. Therefore, increasing cellular MICA levels by preventing those shedding and release may contribute to the clearance of the pathogenic cells and the prevention of HCC. In this study, we constructed a convenient MICA shedding assay system and, by a comprehensive screening, we identified several compounds which may prevent MICA shedding from the cells. Methods: We constructed lentivirual vector expressing MICA protein tagged with nano-luciferase (nanoLuc) in its N terminal because N terminal of MICA is excreted from the cells by shedding. We expressed this construct in HBV replicating cells, HepG2.2.15, and after adding compounds, we determined the luciferase activities in the supernatant and cell lysates. Results: We picked up the samples which showed low activities in supernatant with high activities in the cell lysates, indicating low levels of shedding and high levels of remaining cellular MICA. Out of over 1,000 compounds screened, we determined four candidate compounds which efficiently prevented MICA shedding. We confirmed the effects of these compounds in other cell systems infected with HBV. Conclusion: Because these compounds enhanced the expression levels of cellular MICA and decreased its shedding in HBV-replicating human hepatocytes, using these compounds may be favorable during HBV chronic infection to enhance the NK cell-mediated clearance of the pathogenic hepatocytes and to prevent HCC. Citation Format: Motoko Ohno, Motoyuki Otsuka, Takahiro Kishikawa, Takeshi Yoshikawa, Akemi Takata, Kazuhiko Koike. Prevention of MICA shedding from HBV infected hepatocytes to activate NK cells for better clearance of malignant foci and virus-infected cells. [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 4456. doi:10.1158/1538-7445.AM2015-4456
    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 ...
  • 9
    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 ...
  • 10
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...