25 research outputs found

    Upregulation of Protein Tyrosine Phosphatase Type IVA Member 3 (PTP4A3/PRL-3) is Associated with Tumor Differentiation and a Poor Prognosis in Human Hepatocellular Carcinoma

    Get PDF
    BACKGROUND: Protein tyrosine phosphatase type IVA member 3 (PTP4A3/PRL-3), a metastasis-associated phosphatase, plays multiple roles in cancer metastasis. We investigated PTP4A3/PRL-3 expression and its correlation with the clinicopathological features and prognosis in hepatocellular carcinoma (HCC). METHODS: Gene expression profiles of PTP4A3/PRL-3 were obtained in poorly differentiated HCC tissues. The results were validated independently by TaqMan gene expression assays and immunohistochemical analysis. RESULTS: According to the microarray profiles, PTP4A3/PRL-3 was upregulated in patients with poorly differentiated disease compared to patients with well-differentiated disease with hepatic backgrounds associated with hepatitis B or C. Validation analysis showed that the PTP4A3/PRL-3 mRNA and protein levels were significantly associated with poor differentiation (PĀ <Ā 0.0001), high serum Ī±-fetoprotein (PĀ <Ā 0.01), high serum protein induced by vitamin K absence/antagonist-II (PIVKA-II), and hepatic vascular invasion (PĀ <Ā 0.05). The expression of PTP4A3/PRL-3 protein was also correlated with advanced cancer stages (PĀ <Ā 0.01); this resulted in a significantly poorer prognosis in both overall (PĀ =Ā 0.0024) and recurrence-free survival (PĀ =Ā 0.0227). According Cox regression univariate analysis, the positive expression of PTP4A3/PRL-3 was a poor risk prognostic factor (OS, PĀ =Ā 0.0031; recurrence-free survival, PĀ =Ā 0.0245). Cox regression multivariate analysis indicated that high PTP4A3/PRL-3 expression was an independent, unfavorable prognostic factor for overall survival (hazard ratio 0.542; PĀ =Ā 0.048). CONCLUSIONS: PTP4A3/PRL-3 might be closely associated with HCC progression, invasion, and metastasis. Its high expression had a negative impact on the prognosis of HCC patients. This strongly suggests that PTP4A3/PRL-3 should be considered as a prognostic factor. Further analysis should be pursued to evaluate it as a novel prognostic target. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1245/s10434-012-2395-2) contains supplementary material, which is available to authorized users

    Anti-Tumor Effects of Second Generation Ī²-Hydroxylase Inhibitors on Cholangiocarcinoma Development and Progression

    Get PDF
    Cholangiocarcinoma (CCA) has a poor prognosis due to widespread intrahepatic spread. Aspartate Ī²-hydroxylase (ASPH) is a transmembrane protein and catalyzes the hydroxylation of aspartyl and asparaginyl residues in calcium binding epidermal growth factor (cbEGF)-like domains of various proteins, including Notch receptors and ligands. ASPH is highly overexpressed (\u3e95%) in human CCA tumors. We explored the molecular mechanisms by which ASPH mediated the CCA malignant phenotype and evaluated the potential of ASPH as a therapeutic target for CCA. The importance of expression and enzymatic activity of ASPH for CCA growth and progression was examined using shRNA ā€œknockdownā€ and a mutant construct that reduced its catalytic activity. Second generation small molecule inhibitors (SMIs) of Ī²-hydroxylase activity were developed and used to target ASPH in vitro and in vivo. Subcutaneous and intrahepatic xenograft rodent models were employed to determine anti-tumor effects on CCA growth and development. It was found that the enzymatic activity of ASPH was critical for mediating CCA progression, as well as inhibiting apoptosis. Mechanistically, ASPH overexpression promoted Notch activation and modulated CCA progression through a Notch1-dependent cyclin D1 pathway. Targeting ASPH with shRNAs or a SMI significantly suppressed CCA growth in vivo

    Aspartate Ī²-Hydroxylase Expression Promotes a Malignant Pancreatic Cellular Phenotype

    Get PDF
    Pancreatic cancer (PC) is one of the leading causes of cancer related deaths due to aggressive progression and metastatic spread. Aspartate Ī²-hydroxylase (ASPH), a cell surface protein that catalyzes the hydroxylation of epidermal growth factor (EGF)-like repeats in Notch receptors and ligands, is highly overexpressed in PC. ASPH upregulation confers a malignant phenotype characterized by enhanced cell proliferation, migration, invasion and colony formation in vitro as well as PC tumor growth in vivo. The transforming properties of ASPH depend on enzymatic activity. ASPH links PC growth factor signaling cascades to Notch activation. A small molecule inhibitor of Ī²-hydroxylase activity was developed and found to reduce PC growth by downregulating the Notch signaling pathway. These findings demonstrate the critical involvement of ASPH in PC growth and progression, provide new insight into the molecular mechanisms leading to tumor development and growth and have important therapeutic implications

    LRH1 as a driving factor in pancreatic cancer growth

    Get PDF
    Liver receptor homolog 1 (LRH1), directs the development and differentiation of embryonic pancreas, and is overexpressed in pancreatic cancer (PC). We hypothesized that LRH1 promotes PC growth. Cell proliferation and tumorigenicity in nude mice were compared between empty vector-transfected (control) and stable LRH1-overexpressed PC cell lines. The subsequent tumor burden, vasculature development, and histologic features were evaluated. LRH1 overexpression enhanced the expression of downstream target genes (cyclin D1/E1) and stimulated cell proliferation in PC cell lines. LRH1 upregulated cyclin E1 truncated T1/T2 isoforms expression which may occur through ERĪ±ā€“calpain1 signaling. Compared with the control, LRH1 overexpressing stable cells generated tumors with increased weight, proliferation index and enhanced angiogenesis. Cyclin D1/E1 and calpain1 were overexpressed in human PC tumors compared to adjacent normal pancreas. These observations demonstrate that LRH1 promotes PC growth and angiogenesis, suggesting that LRH1 is a driving factor in tumorigenesis and may serve as a potential therapeutic target

    Aspartate Ī²-Hydroxylase Expession Promotes a Malignant Pancreatic Cellular Phenotype

    Get PDF
    Pancreatic cancer (PC) is one of the leading causes of cancer related deaths due to aggressive progression and metastatic spread. Aspartate Ī²-hydroxylase (ASPH), a cell surface protein that catalyzes the hydroxylation of epidermal growth factor (EGF)-like repeats in Notch receptors and ligands, is highly overexpressed in PC. ASPH upregulation confers a malignant phenotype characterized by enhanced cell proliferation, migration, invasion and colony formation in vitro as well as PC tumor growth in vivo. The transforming properties of ASPH depend on enzymatic activity. ASPH links PC growth factor signaling cascades to Notch activation. A small molecule inhibitor of Ī²-hydroxylase activity was developed and found to reduce PC growth by downregulating the Notch signaling pathway. These findings demonstrate the critical involvement of ASPH in PC growth and progression, provide new insight into the molecular mechanisms leading to tumor development and growth and have important therapeutic implications

    Proteasome activity is required for the initiation of precancerous pancreatic lesions

    Get PDF
    Proteasome activity is significantly increased in advanced cancers, but its role in cancer initiation is not clear, due to difficulties in monitoring this process in vivo. We established a line of transgenic mice that carried the ZsGreen-degronODC (Gdeg) proteasome reporter to monitor the proteasome activity. In combination with Pdx-1-Cre;LSL-KrasG12D model, proteasome activity was investigated in the initiation of precancerous pancreatic lesions (PanINs). Normal pancreatic acini in Gdeg mice had low proteasome activity. By contrast, proteasome activity was increased in the PanIN lesions that developed in Gdeg;Pdx-1-Cre;LSL-KrasG12D mice. Caerulein administration to Gdeg;Pdx-1-Cre;LSL-KrasG12D mice induced constitutive elevation of proteasome activity in pancreatic tissues and accelerated PanIN formation. The proteasome inhibitor markedly reduced PanIN formation in Gdeg;Pdx-1-Cre;LSL-KrasG12D mice (Pā€‰=ā€‰0.001), whereas it had no effect on PanIN lesions that had already formed. These observations indicated the significance of proteasome activity in the initiation of PanIN but not the maintenance per se. In addition, the expressions of pERK and its downstream factors including cyclin D1, NF-ĪŗB, and Cox2 were decreased after proteasome inhibition in PanINs. Our studies showed activation of proteasome is required specifically for the initiation of PanIN. The roles of proteasome in the early stages of pancreatic carcinogenesis warrant further investigation

    ASPH upregulated the expression of cyclin D1 expression and accelerated cell proliferation.

    No full text
    <p><b>(A)</b> Relative CCND1 mRNA expression in H1, RBE, SSP25, and ETK1 cells infected with lentivirus containing shLuc or shASPH. <b>(B)</b> Immunoblotting of ASPH, cyclin D1, and intracellular domain of Notch1 (ICN) in RBE-shLuc, RBE-shASPH, ETK1-shLuc, and ETK1-shASPH transfected cells with or without overexpression of ICN. <b>(C)</b> Relative cell proliferation in RBE-shLuc, RBE-shASPH, ETK1-shLuc, and ETK1-shASPH in the presence or absence of 10 Ī¼M of the Ī³-secretase inhibitor DAPT using the MTT assay. <b>(D)</b> Relative cell proliferation rate in RBE-shLuc, RBE-shASPH, RBE-shASPH transfected cells overexpressing ICN. ***, <i>p</i> <0.001; **, <i>p</i> <0.01; *, <i>p</i> <0.05.</p

    Dominant Expression of DCLK1 in Human Pancreatic Cancer Stem Cells Accelerates Tumor Invasion and Metastasis

    No full text
    <div><p>Patients with pancreatic cancer typically develop tumor invasion and metastasis in the early stage. These malignant behaviors might be originated from cancer stem cells (CSCs), but the responsible target is less known about invisible CSCs especially for invasion and metastasis. We previously examined the proteasome activity of CSCs and constructed a real-time visualization system for human pancreatic CSCs. In the present study, we found that CSCs were highly metastatic and dominantly localized at the invading tumor margins in a liver metastasis model. Microarray and siRNA screening assays showed that doublecortin-like kinase 1 (DCLK1) was predominantly expressed with histone modification in pancreatic CSCs with invasive and metastatic potential. Overexpression of DCLK1 led to amoeboid morphology, which promotes the migration of pancreatic cancer cells. Knockdown of DCLK1 profoundly suppressed in vivo liver metastasis of pancreatic CSCs. Clinically, DCLK1 was overexpressed in the metastatic tumors in patients with pancreatic cancer. Our studies revealed that DCLK1 is essential for the invasive and metastatic properties of CSCs and may be a promising epigenetic and therapeutic target in human pancreatic cancer.</p></div

    ASPH modulated Notch signaling in CCA cells.

    No full text
    <p><b>(A)</b> Protein expression levels of ASPH, activated Notch1, JAG1, HEY1, and HES1 in 5 CCA cell lines. <b>(B)</b> ASPH, cyclin D1, EpCAM, HES1, HEY1, and cleaved caspase-3 in HEK293 cells transfected with WT-ASPH plasmid at concentrations of 0, 0.5, 1, 1.5, 2, and 2.5 Ī¼g. <b>(C)</b> Semi-quantitation of immunoblotting results depicted in (<b>B</b>). <b>(D)</b> Expression levels of ASPH, cyclin D1, EpCAM, CD44, HES1, HEY1, and cleaved caspase-3 in H1, RBE, and ETK1 cells infected with lentivirus containing either shLuc or shASPH. <b>(E)</b> pCS2-Notch1-full-length-6MT (pCS2-Notch1-F.L.-6MT), EV, ASPH, ASPH<sup>H675Q</sup>, and 12XCSL-DsRedExpressDL (Notch reporter) were co-transfected into HEK293 cells and image of red fluorescence was quantified under a fluorescence microscope. (F) Quantitation of red fluorescence signals are presented. Constitutive active Notch1 (pCS2-Notch1-Ī”EMV-6MT) was used as a positive control. Transfection of Notch reporter construct alone was used as a negative control.</p
    corecore