Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Tumour Biol ; 39(5): 1010428317699120, 2017 May.
Article in English | MEDLINE | ID: mdl-28459212

ABSTRACT

Pancreatic cancer has one of the lowest survival rates of all cancers. The mechanism underlying chemo-resistance of pancreatic cancer is not well understood. Our previous article reported that small molecule YM155 induced apoptosis in pancreatic cancer cells via activation of death receptor 5. In this study, we aim to continuously address death receptor 5-mediated apoptosis in chemo-resistant pancreatic carcinoma. We found that in comparison to paired pancreatic cancer tissues and adjacent normal tissues, five of the six cancer tissues had downregulated death receptor 5 and upregulated Bcl-xL. Mono treatment with lexatumumab was not sufficient to induce apoptosis in pancreatic cancer cells, whereas focal adhesion kinase inhibitor PF573228 significantly sensitized lexatumumab-induced apoptosis. Western blotting analysis revealed that lexatumumab and PF573228 combination treatment increased death receptor 5 but decreased Bcl-xL expression. Interestingly, pre-treatment with Bcl-xL inhibitor ABT263 reversed the insensitivity of panc-1 cells to lexatumumab or PF573228-induced apoptosis. Specific small interfering RNA-mediated gene silencing of Bcl-xL effectively sensitized pancreatic cancer cells to lexatumumab or PF573228-induced apoptosis. Furthermore, lexatumumab and PF573228 combination was shown to exhibit significant xenograft pancreatic tumor growth inhibition in SCID mice. Our data provide fundamental evidence to support the notion that lexatumumab and PF573228 co-treatment could be a potentially effective regime for patients with pancreatic cancer.


Subject(s)
Apoptosis/genetics , Focal Adhesion Protein-Tyrosine Kinases/genetics , Pancreatic Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , bcl-X Protein/genetics , Aniline Compounds/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Quinolones/administration & dosage , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Sulfonamides/administration & dosage , Sulfones/administration & dosage , Xenograft Model Antitumor Assays , bcl-X Protein/antagonists & inhibitors , Pancreatic Neoplasms
2.
Free Radic Biol Med ; 88(Pt B): 212-220, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26279476

ABSTRACT

Intrauterine exposure to gestational diabetes, pre-eclampsia or intrauterine growth restriction alters the redox status of the developing fetus. Such pregnancy-related diseases in most cases do not have a readily identifiable genetic cause, and epigenetic 'priming' mechanisms in utero may predispose both mother and child to later-life onset of cardiovascular and metabolic diseases. The concept of 'fetal programing' or 'developmental priming' and its association with an increased risk of disease in childhood or adulthood has been reviewed extensively. This review focuses on adaptive changes in the in utero redox environment during normal pregnancy and the consequences of alterations in redox control associated with pregnancies characterized by oxidative stress. We evaluate the evidence that the Keap1-Nrf2 pathway is important for protecting the fetus against adverse conditions in utero and may itself be subject to epigenetic priming, potentially contributing to an increased risk of vascular disease and insulin resistance in later life.


Subject(s)
Disease Susceptibility/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology , Animals , Diabetes, Gestational , Female , Fetal Development/physiology , Humans , Oxidation-Reduction , Pregnancy , Pregnancy Complications/metabolism , Prenatal Exposure Delayed Effects/metabolism
3.
Mol Cancer Ther ; 14(1): 80-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25344582

ABSTRACT

Despite much effort, pancreatic cancer survival rates are still dismally low. Novel therapeutics may hold the key to improving survival. YM155 is a small molecule inhibitor that has shown antitumor activity in a number of cancers by reducing the expression of survivin. The aim of our study is to understand the mechanisms by which YM155 functions in pancreatic cancer cells. We established the antitumor effect of YM155 with in vitro studies in cultured cells, and in vivo studies using a mouse xenograft model. Our data demonstrated that YM155 reduced the expression of survivin; however, downregulation of survivin itself is insufficient to induce apoptosis in pancreatic cancer cells. We showed for the first time that treatment with YM155 increased death receptor 5 (DR5) expression in pancreatic cancer cells. We found that YM155 induced apoptosis by broad-spectrum inhibition of IAP family member proteins (e.g., CIAP1/2 and FLIP) and induced proapoptotic Bak protein upregulation and activation; the antitumor effect of YM155 treatment with either the DR5 agonist lexatumumab or gemcitabine on pancreatic cancer cells was synergistic. Our data also revealed that YM155 inhibits tumor growth in vivo, without apparent toxicity to the noncancerous human pancreatic ductal epithelial cell line. Together, these findings suggest that YM155 could be a novel therapeutic agent for pancreatic cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Imidazoles/administration & dosage , Naphthoquinones/administration & dosage , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, SCID , Naphthoquinones/pharmacology , Pancreatic Neoplasms/genetics , Survivin , Xenograft Model Antitumor Assays , Gemcitabine , Pancreatic Neoplasms
4.
Lab Invest ; 93(1): 8-19, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23108376

ABSTRACT

Sorafenib has been used to treat advanced hepatocellular carcinoma (HCC), but the underlying molecular mechanisms remain controversial and why some patients do not respond to this therapy is poorly understood. In this study, we show that sorafenib triggers cell growth inhibition and apoptosis in HCC cells by directly targeting the mitochondria. Treatment with sorafenib induces rapid mitochondrial fragmentation, which is associated with the deregulation of mitochondria fusion-related protein optic atrophy 1 (OPA1). Exposure of cells or isolated mitochondria to sorafenib substantially induces cytochrome c release. Our data indicate that siRNA-mediated OPA1 knockdown significantly sensitizes HCC cells to sorafenib-induced apoptosis. Furthermore, sorafenib has no apparent apoptotic toxicity to normal human primary hepatocytes. Sorafenib inhibits HCC xenograft tumor growth in vivo and murine xenograft tumor tissue analysis reveals mitochondria fusion protein. OPA1 expression levels are strongly downregulated by sorafenib treatment. Western blotting evaluation of patient HCC with matched non-tumor tissue samples demonstrates that OPA1 expression is decreased in up to 40% of HCC patients. Taken together, we have shown that sorafenib suppresses the tumorigenesis of HCC through the induction of mitochondrial injury via OPA1. Our results provide new insights into the pathogenesis of HCC and suggest that OPA1 is a novel therapeutic target in patients with HCC.


Subject(s)
Apoptosis/drug effects , GTP Phosphohydrolases/metabolism , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Animals , Apoptosis/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cytochromes c/metabolism , Down-Regulation/drug effects , GTP Phosphohydrolases/genetics , Gene Knockdown Techniques , Humans , Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, SCID , Mitochondria/drug effects , Mitochondria/pathology , Niacinamide/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Sorafenib , Xenograft Model Antitumor Assays , raf Kinases/metabolism , ras Proteins/metabolism
5.
Clin Chim Acta ; 400(1-2): 107-10, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19000666

ABSTRACT

BACKGROUND: Genomic DNA sequences in cell-free plasma are biomarkers of cancer prognosis, where characteristic changes in methylation of tumour suppressor or oncogene DNA regions are indicative of changes in gene activity. Also, cell-free fetal DNA can be distinguished, by its methylation status, from the maternal DNA in the plasma of pregnant women, hence providing DNA biomarkers for the proposed minimally-invasive diagnosis of fetal aneuploidies, including Down's syndrome. However, the production and clearance of cell-free DNA from plasma in relation to its methylation status, are poorly understood processes. METHODS: We studied the methylation status of DNA derived from the imprinted GNAS1 locus, in cell-free plasma DNA of healthy adults. Heterozygotes were identified that carried the SNP rs1800905 in the imprinted region. The parent-of-origin-dependent DNA methylation was analysed by bisulfite conversion, followed by cloning and sequencing. RESULTS: Genomic DNA molecules derived from both the methylated, maternal, allele and the unmethylated, paternal, allele were found in plasma. Methylated and unmethylated DNA molecules were present in equal numbers. CONCLUSIONS: Our data indicate that the methylation status of a DNA sequence has no effect on its steady state concentration in the cell-free DNA component of plasma, in healthy adults.


Subject(s)
DNA Methylation , DNA/blood , DNA/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Genomic Imprinting , Adult , Base Sequence , Cell-Free System , Chromogranins , Chromosomes, Human, Pair 20/genetics , Genome, Human/genetics , Humans , Molecular Sequence Data
6.
Clin Biochem ; 42(7-8): 736-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19083998

ABSTRACT

OBJECTIVES: To assess whether different genomic cell-free DNAs are equally abundant in the plasma of individual donors, and any relationship between DNA methylation and representation in plasma. DESIGN AND METHODS: The concentrations of DNA in plasma were determined by real-time PCR. RESULTS: Different DNA sequences were not equally represented. The relative abundances were similar in different donors. CONCLUSIONS: Different DNA sequences are not equally abundant in plasma, with no relationship between DNA methylation and abundance.


Subject(s)
DNA/blood , DNA/chemistry , Base Sequence , Humans , Polymerase Chain Reaction , Sequence Analysis, DNA
7.
Prenat Diagn ; 28(1): 1-6, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18022821

ABSTRACT

The discovery of cell-free fetal (cff) DNA and RNA in the maternal circulation has driven developments in noninvasive prenatal diagnosis (NIPD) for the past decade. Detection of paternally derived alleles in cff DNA is becoming well established. Now much interest is focussing on NIPD of fetal chromosomal abnormalities, such as trisomy 21, which is a considerable challenge because this demands accurate quantitative measurements of the amounts of specific cff DNA or cff RNA sequences in maternal blood samples. Emerging strategies for distinguishing and quantifying the fetal nucleic acids in the maternal circulation promise continued development of the field, and pose a number of unanswered questions.


Subject(s)
Aneuploidy , DNA/blood , Prenatal Diagnosis/methods , RNA/blood , Down Syndrome/diagnosis , Down Syndrome/genetics , Female , Fetus/chemistry , Humans , Pregnancy , Trisomy/diagnosis , Trisomy/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...