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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 5200-5200
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 5200-5200
    Abstract: Introduction: Nestin, a class VI intermediate filament, is expressed in pancreatic exocrine progenitor cells. Previously, we reported that 30% of PDAC cases expressed nestin in cancer cells, and its expression was correlated with invasion (Kawamoto, et al. Hum Pathol 2009). Knockdown of nestin using shRNA in human PDAC cell lines inhibited cell migration and invasion in vitro and liver metastasis in vivo. Restoring the expression level of nestin returned the inhibition of cell migration and invasion to the previous level (Matsuda, et al. Cancer Biol Ther 2011). In this study we analyzed the expression and roles of nestin in pancreatic cancer stem cells. Methods: Expressions of stem cell markers and nestin were analyzed using immunocytochemistry and real-time PCR. To assess cancer stem cells, a sphere formation assay was performed as follows: cells were grown in an ultra-low attachment surface plate supplemented with epithelial growth factor and fibroblast growth factor 2. Nestin-regulating molecules were clarified by DNA microarray analysis. Results: The expression level of nestin was high in sphere-forming PDAC cells, and its expression levels correlated with sphere-forming ability. In DNA microarray analysis using nestin shRNA-transfected clones and nestin-expression vector transfected-clones, at a 2-fold cut-off, only transcriptional factor 4 (TCF4) showed a compatible change coincident with nestin expression. It has been reported that TCF4 is involved in the Wnt pathway, and negatively regulates the transcription of NANOG. In real-time PCR, expression levels of TCF4 and NANOG correlated with nestin expression level. Conclusion: Nestin expression levels correlated with sphere-forming ability and expression levels of TCF4 and NANOG. These results indicate that nestin regulate stemness via TCF4 and NANOG.The expression of nestin is associated with highly metastatic cancer stem-like cells, and nestin regulates migration, invasion, and metastasis by altering the pancreatic cancer stem cell function. 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 5200. doi:1538-7445.AM2012-5200
    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 ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 285-285
    Abstract: Backgrounds: Colorectal carcinoma (CRC) is one of the most common cancers throughout the world, and the prognosis remains unfavorable when the disease has progressed to an unresectable stage. New therapeutic strategies such as molecular-targeted agent are a high priority for advanced CRC. Single nucleotide polymorphisms of Fibroblast growth factor receptor 2 (FGFR2) gene are associated with breast and endometrial carcinogenesis. Therefore, FGFR2 has been noted as a novel candidate for molecular targeting of certain cancers. Alternative splicing variants of FGFR2, IIIb and IIIc, determine the binding specificity of fibroblast growth factor (FGF) members, and class switch of FGFR2IIIb to FGFR2IIIc is associated with a more malignant phenotype in prostate and bladder cancers. In CRCs, we have previously reported that overexpression of FGFR2IIIb was associated with the well-differentiated histological type, and expression of FGF7, a specific ligand for FGFR2IIIb was related to angiogenesis and adhesion to type-IV collagen. However, the expression and role of FGFR2IIIc in CRC have not been well clarified. In the present study, we examine the expression and roles of FGFR2IIIc mRNA and proteins in CRC cell lines and human CRC tissues. Methods: To determine the expression of FGFR2IIIc, we used immunohistochemical staining (IHC) and in situ hybridization analysis (ISH) in human colorectal tissues; the tissues were diagnosed as hyperplastic polyp, adenoma, adenocarcinoma and carcinoma in/with adenoma, according to the criteria of the World Health Organization. Furthermore, we constructed a FGFR2 IIIc-gene-transfected CRC cell line from DLD-1cells and FGFR2IIIc- shRNA-transfected CRC cell line from LoVo cells, and examined the effects of FGFR2 IIIc on cell behaviors. Results: In normal colorectal tissues, IHC and ISH showed the expression of FGFR2IIIc in fibroblasts, endothelium and the surface layer of normal colorectal epithelium. The expression level of FGFR2IIIc was increased in the following order; carcinoma & gt; adenoma & gt; hyperplastic polyp. In CRC cases, expression of FGFR2IIIc correlates with liver metastasis and poor prognosis. FGFR2IIIc-stably transfected DLD-1 cells, which overexpress FGFR2 IIIc, induced increases in colony-forming activities. In contrast, FGFR2IIIc shRNA-stably transfected LoVo cells, which express a lower level than sham transfected cells, induced decreases in colony-forming activities. Heterotopic and orthotopic implantation of FGFR2 IIIc-transfected DLD-1 cells in nude mice increased the tumor volume compared to that in mice receiving sham-transfected cells. Conclusion: These findings suggest that FGFR2IIIc plays important roles in colorectal carcinogenesis as well as tumor progression. FGFR2IIIc may thus serve as a novel candidate for molecular targeting in CRCs. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 285.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    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: Journal of Histochemistry & Cytochemistry, SAGE Publications, Vol. 59, No. 1 ( 2011-01), p. 68-75
    Abstract: Xenograft transplantation of human tumor cells into immunodeficient mice is an important method to clarify the roles of specific molecules or chemicals in vivo. Recently, this method has been reported as a definitive examination to identify tumor stem cells. In this study, the authors compared the morphology and the quality and quantity of ribonucleic acid (RNA) and protein in paraffin-embedded tissues of nude mice implanted with human uterine cervical cancer cells, followed by fixation with commonly used fixatives, including 4% paraformaldehyde (PFA), 10% neutral buffered formalin (NBF), 20% NBF, and 99% ethanol (EtOH). The quality of the isolated RNA from PFA- and NBF-fixed paraffin-embedded tissues was high, while EtOH-fixed tissues showed degradation of RNA. NBF-fixed tissues showed excellent quality of morphology, but EtOH-fixed tissues showed contraction of cells. Immunohistochemical results showed differences depending on fixations. The 99% EtOH-fixed samples showed decreases of Ki-67 and VEGF-A immunoreactivities, but improved cytokeratin immunoreactivity. This study indicated that formalin fixation is better than alcohol fixation for RNA preservation in paraffin-embedded cancer cell implantation models. Immunohistochemical results differed markedly depending on fixation materials and antibodies; therefore, suitable fixations are needed to quantify and compare the results of immunohistochemical staining on cancer cell implanted nude mice tissues.
    Type of Medium: Online Resource
    ISSN: 0022-1554 , 1551-5044
    Language: English
    Publisher: SAGE Publications
    Publication Date: 2011
    detail.hit.zdb_id: 1421306-0
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 249-249
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 249-249
    Abstract: Introduction: Nestin is a member of the class VI intermediate filaments and it was first described as a central nervous system (CNS) progenitor/stem cell marker. Nestin is highly expressed in various cancers and was recently reported as a tumor stem cell marker for glioblastoma and melanoma. Previously,we have reported that 30% of pancreatic cancer cases expressed nestin in cancer cells, and its expression correlated with nerve invasion and the presence of cancer cells in the tumor resection margins (Kawamoto, et al. Hum Pathol 2009). Knockdown of nestin exhibited a sheet-like appearance with tight cell-cell adhesion, increased expression of filamentous actin and E-cadherin, and attenuated migration and invasion (Matsuda, et al. Cancer Biol Ther 2011). These findings suggest that nestin is a novel therapeutic target for pancreatic cancer. In this study, we examined the effects of siRNA targeting nestin on pancreatic cancer cell growth migration, and invasion in vitro and in vivo. Methods: Two types of siRNA targeting nestin, which binds to different sites of nestin mRNA, were prepared and transfected to PANC-1 and PK-45H cells. Cell growth, migration, invasion and sphere formation abilities of these cells were analyzed in vitro. We also determined the synergistic effects of gemcitabine in pancreatic cancer cells under treatment with siRNA targeting nestin. For an in vivo study, we constructed luciferase-gene-transfected pancreatic cancer cells and orthotopically implanted the cells into the pancreas of NOD/Shi-scid/IL-2Rγnull (NOG) mice. We injected siRNA into the tail vein of NOG mice once per week with Invivofectamine. Every week, luciferin was injected into the peritoneal cavity of NOG mice and images of primary and metastases were taken using IVIS. After 6 weeks, animals were sacrificed, and the primary and metastatic tumors were macro and microscopically examined. Results: siRNA targeting nestin inhibited the growth, migration, invasion and sphere-forming ability of both PANC-1 and PK-45H cells. Treatment with gemcitabine, in addition to the treatment with siRNA targeting nestin in pancreatic cancer cells, decreased cell viability as compared with only gemcitabine, siRNA targeting nestin or control siRNA- treated pancreatic cancer cells. In the orthotopic implantation model, siRNA targeting nestin significantly decreased primary tumor formation in the pancreas, as compared with negative control siRNA groups. Furthermore, nestin siRNA markedly suppressed the formation of metastatic nodules in the liver and lungs. Conclusion: In pancreatic cancer cells, siRNA targeting nestin inhibited cell growth, migration, invasion and sphere formation in vitro, and primary tumor growth and metastatic tumor formation in vivo. Furthermore, siRNA targeting nestin had synergistic effects with gemcitabine. Inhibition of nestin using siRNA may be a novel therapeutic optiont for pancreatic cancer treatment. Citation Format: Toshiyuki Ishiwata, Hisashi Yoshimura, Taeko Suzuki, Yuji Yanagisawa, Yoko Kawamoto, Kiyoko Kawahara, Zenya Naito, Murray Korc, Yoko Matsuda. Inhibition of nestin using siRNA as a novel therapeutic option for pancreatic cancer. [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 249. doi:10.1158/1538-7445.AM2013-249
    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 ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 3750-3750
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 3750-3750
    Abstract: Background: Nestin is a class VI intermediate filament, first described as a central nervous system (CNS) progenitor/stem cell marker and recently reported as a tumor stem cell marker in several malignant tumors, including glioblastoma and melanoma. In the normal pancreas, nestin is expressed in exocrine progenitor cells. Previously, we have reported that 30% of pancreatic cancer cases expressed nestin in cancer cells, and its expression correlated with nerve invasion and the presence of cancer cells in the tumor resection margins (Kawamoto, et al. Hum Pathol 2009). Knockdown of nestin exhibited a sheet-like appearance with tight cell-cell adhesion, increased expression of filamentous-actin and E-cadherin, and attenuated migration and invasion, both of which recovered following nestin re-expression (Matsuda, et al. Cancer Biol Ther 2011). Furthermore, knockdown of nestin suppressed sphere-forming ability, and altered the expression levels of stemness-related genes such as NANOG and transcription factor 4 (Matsuda, et al. AACR 2012). Therefore, nestin is considered to be a novel therapeutic target for pancreatic cancer via suppression of cell migration, invasion, metastasis and stemness. To develop nestin-targeting therapy, we focused on the phosphorylation of nestin protein. Phosphorylation of nestin at two different sites of progenitor cells of the CNS and myocytes has been reported to regulate the cell cycle via activation of cyclin-dependent kinase 5 and cdc2 kinase (Sahlgren, et al. Mol Cell Biol 2003). In this study, we analyzed the expression and roles of nestin phosphorylation in pancreatic cancer cells. Methods: Expression levels of phosphorylated nestin in pancreatic cancer cells were determined by flow cytometer, fluorescent staining and Western blot analyses. To assess the functions of phosphorylated nestin, we prepared three mutated nestin expression vectors which possess mutations at two different phosphorylated sites, both in nestin. A human pancreatic cancer cell line, MIA PaCa-2, which express very low levels of nestin was transfected with the mutated or wild-type nestin-expression vector. Results: Expression level of phosphorylated nestin was higher in M-phase cells than S- or G0/G1-phase cells. Phosphorylated nestin was strongly and diffusely localized in M-phase cells, while it was weakly detected in the nuclei of cells in other phases. Mutated nestin in phosphorylated site-transfected cells showed lower levels of cell growth, migration and invasion than wild-type nestin-transfected cells. Furthermore, mutated nestin-transfected cells had a tendency towards increasesed multinuclear cells. Conclusion: Phosphorylation of nestin affects the cell growth, migration and invasion of pancreatic cancer cells, and phosphorylated nestin may play important roles in the regulation of the cell cycle. Inhibition of phosphorylated nestin may be a novel target for pancreatic cancer treatment. Citation Format: Yoko Matsuda, Hisashi Yoshimura, Taeko Suzuki, Zenya Naito, Kiyoko Kawahara, Yuji Yanagisawa, Yoko Kawamoto, Toshiyuki Ishiwata. Inhibition of phosphorylation of nestin decrease pancreatic cancer cell growth. [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 3750. doi:10.1158/1538-7445.AM2013-3750
    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 ...
  • 6
    In: Cancer Science, Wiley, Vol. 105, No. 9 ( 2014-09), p. 1212-1219
    Abstract: The alternative splicing of the extracellular domain of fibroblast growth factor receptor ( FGFR )‐2 generates the III b and III c isoforms. Expression of FGFR ‐2 III b correlates with vascular endothelial growth factor‐A ( VEGF ‐A) expression and venous invasion of pancreatic ductal adenocarcinoma ( PDAC ). By contrast, FGFR ‐2 III c expression correlates with faster development of liver metastasis after surgery, and increased proliferation rates and invasion of the cancer. In this study, we analyzed the expression and roles of total FGFR ‐2 (both isoforms) to determine the effectiveness of FGFR ‐2‐targeting therapy for PDAC . Immunohistochemically, FGFR ‐2 was highly expressed in 25/48 (52.1%) PDAC cases, and correlated with advanced stage cancer. In FISH analysis, FGFR 2 was amplified in 3/7 PDAC cell lines. We stably transfected an FGFR ‐2 sh RNA targeting the III b and III c isoforms into FGFR 2 ‐amplified PDAC cells. The proliferation rates, migration, and invasion of FGFR ‐2‐sh RNA ‐transfected cells were lower than those of control cells in vitro . In response to FGF ‐2, FGFR ‐2‐sh RNA ‐transfected cells showed decreased phosphorylation of ERK compared with control cells. The FGFR ‐2‐sh RNA ‐transfected cells also expressed lower levels of vascular endothelial growth factor‐A than control cells, and formed smaller s.c. tumors in nude mice. These findings suggest that FGFR ‐2 is a therapeutic target for inhibition in PDAC .
    Type of Medium: Online Resource
    ISSN: 1347-9032 , 1349-7006
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2014
    detail.hit.zdb_id: 2115647-5
    detail.hit.zdb_id: 2111204-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 5203-5203
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 5203-5203
    Abstract: Long non-coding RNAs (lncRNAs), non-protein coding transcripts longer than 200 nucleotides, have been recently reported to play important roles in carcinogenesis and cancer metastasis through epigenetic regulation. In the present study, we examined the expression levels and roles of lncRNA in pancreatic cancer to elucidate whether lncRNA could be a novel candidate for pancreatic cancer therapy. First, we injected PANC-1 and PK-45H, pancreatic ductal adenocarcinoma cells into the spleens of NOD/Shi-scid, IL-2Rγnull (NOG) mice, and then established novel cell lines from liver and lung metastatic nodules. Microarray analysis revealed that H19, a member of lncRNA, and CTAG1A and CTAG2, cancer-testis antigens, were the most increased RNAs in PANC-lung-1A cells from lung metastasis as compared to the parental cells (82.4, 84.4 and 62.2-fold, respectively). Quantitative RT-PCR (qRT-PCR) confirmed higher levels of H19 mRNA in the metastatic cell lines from PANC-1 and PK-45H cells than in PANC-Skin cells from subcutaneous tumors. H19 is an imprinted non-coding RNA, and the H19 gene produces a 2.3 kb lncRNA during embryogenesis. Its expression is low or non-existent in normal human tissues. H19 is abundantly expressed in liver, breast, endometrial and bladder cancers, but the roles of H19 that have been reported in these cancers are controversial. In six of 10 pancreatic carcinoma cell lines, the H19 levels determined by qRT-PCR were higher than those of immortalized pancreatic ductal epithelial cell lines (HPDE 4 and 6). Established pancreatic cancer cells from liver metastasis of the same patient (PK-45H) showed higher H19 mRNA levels than those from primary tumor (PK-45P). In situ hybridization analysis using branched DNA probe for H19 mRNA showed that H19 was expressed in ductal cells, but not in acinar and islet cells in normal pancreatic tissues. On the other hand, H19 was strongly expressed in pancreatic cancer cells in 6 out of 38 (16%) pancreatic ductal adenocarcinoma tissues. siRNA targeting H19 was transfected to PANC-lung-1A cells to clarify the roles of H19 in the metastatic cells. The cell proliferation rate was not altered in the siRNA transfected metastatic cells, but cell migration, as observed using a Boyden chamber assay, and sphere formation were significantly decreased in the cells. H19 mRNA in PANC-1 cells was more abundantly detected in the spheres than the non-sphere cells. These findings suggest that H19 lncRNA plays important roles in the metastasis and cancer stem cell functions of pancreatic cancer. H19 is a novel candidate as a therapeutic target for pancreatic cancer metastasis. Note: This abstract was not presented at the meeting. Citation Format: Hisashi Yoshimura, Yoko Matsuda, Taeko Suzuki, Zenya Naito, Toshiyuki Ishiwata. Long non-coding RNA H19 as a novel therapeutic target for pancreatic cancer. [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 5203. doi:10.1158/1538-7445.AM2014-5203
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 1421-1421
    Abstract: BACKGROUND: Nestin, a class VI intermediate filament protein, was first described as a neural stem cell/progenitor cell marker expressed during central nervous system development. Recently, nestin was detected in various neoplasms such as glioblastomas, gastrointestinal stromal tumors, prostate cancers, breast cancers, pancreatic cancers and malignant melanomas. An abundance of nestin has been identified in metastatic sites and the dermal parts of nodular melanomas, and the expression level of nestin has relevance to poor prognosis in malignant melanomas. These findings suggest that nestin plays important roles in the invasion of melanoma cells. Furthermore, recent studies have shown that nestin is one of the cancer stem cell markers in melanomas. Therefore, we hypothesized that nestin may be a novel therapeutic target for malignant melanomas. In this study, we used a silencing strategy to clarify the effectiveness of nestin-targeting therapy in malignant melanomas. METHODS: To determine the expression patterns of nestin, we performed immunohistochemical analysis of nestin in human melanoma tissues. Next, we stably knocked down nestin expression in A375 human malignant melanoma cells by short hairpin RNA (shRNA) transfection in vitro. We also transfected sham vectors to A375 cells as negative controls. Cell proliferation, invasion, morphology, and sphere-forming ability were compared with nestin-shRNA-transfected cells (Sh) and sham cells (Sc). RESULTS: Immunohistochemical staining demonstrated that nestin was strongly expressed in melanoma cells in the invasive front of human melanoma tissues. Reduced expression level of nestin mRNA and protein in Sh cells was confirmed by quantitative RT-PCR, western blot and immunocytochemical analyses. Sh and Sc cells did not show characteristic morphological differences; however, stress fiber formation of F-actin was markedly increased in Sh cells. Cell growth of Sh cells was slower than that of Sc cells at 48 hrs as determined by cell counts and MTT assay. On the cell invasion assay using a modified Boyden chamber coated with matrigel on the inner surface of the inserts, the invasion of Sh cells was significantly attenuated in comparison with that of Sc cells. Sphere-forming ability using ultra-low attachment plates supplemented with EGF and FGF-2 showed that Sh cells formed fewer spheres than Sc cells. CONCLUSION: Nestin is expressed in melanoma cells at the invasive front of melanoma tissues, and decreased expression level of nestin inhibits the growth and invasion of melanoma cells. Furthermore, nestin regulates the functions of cancer stem cells of malignant melanoma as determined by sphere-forming ability. Our findings imply that nestin is a promising novel therapeutic target in malignant melanomas. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 1421. doi:10.1158/1538-7445.AM2011-1421
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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. 71, No. 8_Supplement ( 2011-04-15), p. 5240-5240
    Abstract: BACKGROUND: Pancreatic cancer is associated with extremely high mortality rates due to rapid progression and a high incidence of metastases. Liver and lung metastases in the early postoperative period are one of the causes for the poor prognosis of patients with resected pancreatic cancer. Information on the pathobiology of metastatic pancreatic cancer and elucidation of the underlying molecular mechanisms are urgently needed in order to develop novel and effective therapeutic approaches to treatment. Injection of human pancreatic cancer cells in the spleens of nude mice is one of the major liver metastasis models. However, this model yields few metastatic tumors in the livers and none in the lungs. NOG mice have severe immunodeficiency, including defects of T, B and NK cell functions, and dysfunction of dendritic cells. Human pancreatic cancer cells are reported to easily proliferate and metastasize to other organs in NOG mice. In this study, we established and characterized human pancreatic cancer cell lines from liver and lung metastases in NOG mice. METHODS: PANC-1 cells (1×105 cells) were injected into the spleens of NOG mice. The mice were euthanized 8 weeks after injection, and the livers and lungs were removed. Metastatic tumors of the liver and lung were cut into small pieces and dispersed into single cells in a medium containing antibiotics. Biological and morphological characteristics, and gene expression patterns of mesenchymal markers of the liver and lung metastatic cells, were compared with parental cells. RESULTS: These metastatic cells were confirmed as being of human-cell origin using PCR with primer pair of human mitochondrial DNA. There were no morphological changes between liver and lung metastatic cells and parental cells, but the metastatic cells proliferated more slowly than parental cells as determined by MTT assays and cell counts. By contrast, the metastatic cells exhibited greater migration than the parental cells, as determined in Boyden chamber assays and by time lapse analysis. Attachment assays revealed that adhesion of metastatic cells to the type I and IV collagen, laminin and fibronectin was stronger than that of parental cells. Nestin, a cancer stem cell marker, was highly expressed in the liver and lung metastatic cells. CONCLUSION: These findings suggest that human pancreatic cancer cell lines derived from liver and lung metastases in NOG mice have different characteristics from parental cells. These new cell lines may contribute to resolving the mechanisms of metastasis of pancreatic cancer and the roles of cancer stem cells in metastasis. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 5240. doi:10.1158/1538-7445.AM2011-5240
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 412-412
    Abstract: Background: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a high incidence of distant metastasis. Recent studies have shown that cancer stem cells (CSCs) are important in cancer cell growth, invasion, metastasis, and recurrence. Several studies have revealed some CSC-specific markers for PDAC, such as CD133, CD24, CD44, CXCR4, ESA, and nestin. Some evidence also suggests that PDAC progresses through a multistep process comprised of noninvasive precursor lesions known as pancreatic intraepithelial neoplasias (PanINs). The reports of CSCs in PanINs are limited. We performed a comprehensive analysis of the expression of CSC markers in PDACs, PanINs, and normal pancreatic tissues. Materials & Methods: CD24, CD44, ESA, CD133, CXCR4, and nestin were used as CSC markers. Human PDACs (n=67), PanIN-1 (n=35), PanIN-2 (n=52), PanIN-3 (n=18), and normal pancreatic tissues (n=54), which were obtained from patients who underwent surgical operations in the surgical department of Nippon Medical School, were used for immunohistochemical analysis. Human PDAC cell lines, PANC-1, KLM-1, and MIA PaCa-2, were used for flow cytometric assays and quantitative RT-PCR analysis. Results: Regarding the clinicopathological features, the positivity of CD44 and CD133 showed significant correlations for UICC classifications and histological features In the survival analysis, CD133-positive PDACs led to a significantly poorer prognosis. In the immunohistochemical analysis, CD24, CD44, ESA, CXCR4, and nestin showed a gradual increase of positivity along with the grades of PanINs. Flow cytometric assays and RT-PCR analysis confirmed the expression profile in PDAC. Conclusion: CSC markers were expressed to various extents in PDAC, and the expression of CD133 and CD44 was correlated with the prognosis and stage. Furthermore, CSC markers showed a gradual increase along with the grades of PanINs; therefore, CSC may be involved in the carcinogenesis of PDAC. 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 412. doi:1538-7445.AM2012-412
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...