Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
FEBS J ; 290(16): 4126-4144, 2023 08.
Article in English | MEDLINE | ID: mdl-37095740

ABSTRACT

Our investigation to explore cellular alterations related to undernutrition in cancer cells revealed that the protein level of heterogenous nuclear ribonucleoprotein A1 (hnRNP A1) is drastically decreased by serum/glucose starvation. Its loss was reversible, serum/glucose starvation-specific and universal throughout cell types and species. The hnRNP A1 mRNA level and hnRNP A1 mRNA/protein stability were not altered under this condition. CCND1 mRNA, which we newly identified as the binding target of hnRNP A1, was decreased by serum/glucose starvation. Under similar conditions, CCND1 protein was reduced in vitro and in vivo, whereas hnRNP A1 mRNA level and CCND1 mRNA level revealed no correlation in most clinical samples. Functional analyses revealed that CCND1 mRNA stability is certainly dependent on hnRNP A1 protein level and that RNA recognition motif-1 (RRM1) in hnRNP A1 plays a central role in maintaining CCND1 mRNA stability and subsequent protein expression. The injection of RRM1-deleted hnRNP A1-expressing cancer cells in the mouse xenograft model did not form any tumours, and that of hnRNP A1-expressing cancer cells retained CCND1 expression at the lesion adjacent to necrosis with a slight increase in tumour volume. Furthermore, RRM1 deletion caused growth suppression with the induction of apoptosis and autophagy, whereas CCND1 restoration completely recovered it. Our results indicate that serum/glucose starvation triggers entire hnRNP A1 protein loss, and its loss may play a role in CCND1 mRNA destabilization and CCND1-mediated cellular event inhibition, i.e. growth promotion, apoptosis induction and autophagosome formation.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Humans , Animals , Mice , Heterogeneous Nuclear Ribonucleoprotein A1/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Cyclin D1/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Glucose
2.
Noncoding RNA ; 8(4)2022 Jul 28.
Article in English | MEDLINE | ID: mdl-36005825

ABSTRACT

Virus-encoded microRNAs (miRNAs) target viral and host mRNAs to repress protein production from viral and host genes, and regulate viral persistence, cell transformation, and evasion of the immune system. The present study demonstrated that simian virus 40 (SV40)-encoded miRNA miR-S1 targets a cellular miRNA miR-1266 to derepress their respective target proteins, namely, T antigens (Tags) and telomerase reverse transcriptase (TERT). An in silico search for cellular miRNAs to interact with viral miR-S1 yielded nine potential miRNAs, five of which, including miR-1266, were found to interact with miR-S1 in dual-luciferase tests employing reporter plasmids containing the miRNA sequences with miR-S1. Intracellular bindings of miR-1266 to miR-S1 were also verified by the pull-down assay. These miRNAs were recruited into the Ago2-associated RNA-induced silencing complex. Intracellular coexpression of miR-S1 with miR-1266 abrogated the downregulation of TERT and decrease in telomerase activity induced by miR-1266. These effects of miR-S1 were also observed in miR-1266-expressing A549 cells infected with SV40. Moreover, the infected cells contained more Tag, replicated more viral DNA, and released more viral particles than control A549 cells infected with SV40, indicating that miR-S1-induced Tag downregulation was antagonized by miR-1266. Collectively, the present results revealed an interplay of viral and cellular miRNAs to sequester each other from their respective targets. This is a novel mechanism for viruses to manipulate the expression of viral and cellular proteins, contributing to not only viral lytic and latent replication but also cell transformation observed in viral infectious diseases including oncogenesis.

3.
Biol Pharm Bull ; 43(11): 1715-1728, 2020.
Article in English | MEDLINE | ID: mdl-33132317

ABSTRACT

SV40-encoded microRNA (miRNA), miR-S1, downregulates the large and small T antigens (LTag and STag), which promote viral replication and cellular transformation, thereby presumably impairing LTag and STag functions essential for the viral life cycle. To explore the functional significance of miR-S1-mediated downregulation of LTag and STag as well as the functional roles of miR-S1, we evaluated viral DNA replication and proinflammatory cytokine induction in cells transfected with simian virus 40 (SV40) genome plasmid and its mutated form lacking miR-S1 expression. The SV40 genome encodes two mature miR-S1s, miR-S1-3p and miR-S1-5p, of which miR-S1-3p is the predominantly expressed form. MiR-S1-3p exerted strong repressive effects on a reporter containing full-length sequence complementarity, but only marginal effect on one harboring a sequence complementary to its seed sequence. Consistently, miR-S1-3p downregulated LTag and STag transcripts with complete sequence complementarity through miR-S1-3p-Ago2-mediated mRNA decay. Transfection of SV40 plasmid induced higher DNA replication and lower LTag and STag transcripts in most of the examined cells compared to that miR-S1-deficient SV40 plasmid. However, miR-S1 itself did not affect DNA replication without the downregulation of LTag transcripts. Both LTag and STag induced the expression of tumor necrosis factor α (TNFα) and interleukin (IL)-17F, which was slightly reduced by miR-S1 due to miR-S1-mediated downregulation of LTag and STag. Forced miR-S1 expression did not affect TNFα expression, but increased IL-17F expression. Overall, our findings suggest that miR-S1-3p is a latent modifier of LTag and STag functions, ensuring efficient viral replication and attenuating cytokine expression detrimental to the viral life cycle.


Subject(s)
Antigens, Viral, Tumor/genetics , Gene Expression Regulation, Viral/immunology , MicroRNAs/metabolism , RNA, Viral/metabolism , Simian virus 40/genetics , A549 Cells , DNA Replication/immunology , DNA, Viral/biosynthesis , HEK293 Cells , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Interleukin-17/metabolism , Interleukin-8/metabolism , Polyomavirus Infections/genetics , Polyomavirus Infections/immunology , Polyomavirus Infections/virology , Simian virus 40/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Virus Infections/genetics , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Virus Replication/immunology
4.
Biochem Biophys Res Commun ; 516(2): 388-396, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31217077

ABSTRACT

Inhibition of prostaglandin E2 signaling via EP2/EP4 prostanoid receptors suppresses Insulin-like growth factor (IGF)-1-induced proliferation of pancreatic cancer BxPC-3 cells. To better understand the mechanism of EP2/EP4 signaling for controlling cell proliferation, we performed metabolome analyses in BxPC-3 cells treated with IGF-1 alone or IGF-1 plus EP2/EP4 inhibitors. These analyses revealed increased g-aminobutyric acid and 5-oxoproline production following the addition of EP2/EP4 inhibitors to IGF-1-treated cells. The expression of a 5-oxoproline-catalyzing enzyme, γ-glutamylcyclotransferase (GGCT), was also upregulated by IGF-1 treatment and further enhanced by the addition of EP2/EP4 inhibitors. Knockdown of GGCT expression resulted in the loss of suppressive effects of EP2/EP4 inhibitors on IGF-1-induced BxPC-3 cell proliferation, whereas GGCT overexpression repressed the basal proliferation of BxPC-3 cells but did not affect the suppressive effects of EP2/EP4 inhibitors. To summarize, we propose a role for EP2/EP4 signaling in regulating IGF-1-induced cell proliferation, in which EP2/EP4 signaling represses IGF-1-induced GGCT expression, which mediates and whose amount controls a branch of IGF-1 signaling to promote cell proliferation via extracellular signal-regulated kinase phosphorylation.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction , Up-Regulation , gamma-Glutamylcyclotransferase/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Isoindoles/pharmacology , Metabolome , Pyrrolidonecarboxylic Acid/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Xanthones/pharmacology , gamma-Aminobutyric Acid/metabolism
5.
Hepatol Int ; 12(3): 254-261, 2018 May.
Article in English | MEDLINE | ID: mdl-29651702

ABSTRACT

BACKGROUND AND AIMS: Tsumura-Suzuki obese diabetic (TSOD) is a good model of metabolic syndrome showing typical lesions found in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis, and develops spontaneous hepatic tumors with a high frequency. Majority of the developing tumors overexpress glutamine synthetase (GS), which is used as a marker of hepatocellular carcinoma (HCC). The aim of this study is to assess the status of expression of metabolism-related genes and the level of bile acids in the TSOD mice-derived tumors and to determine the association with metabolic dysregulation between human HCC and TSOD mice-derived tumors. METHODS: GS-positive hepatic tumors or adjacent normal tissues from 71-week-old male TSOD mice were subjected to immunohistochemical staining, quantitative RT-PCR (qRT-PCR), quantitation of cholic acid and taurocholic acid. RESULTS: We found that downregulation of the rate-limiting enzyme for betaine synthesis (BADH), at both mRNA and protein levels in GS-positive TSOD mice-derived tumors. Furthermore, the bile acid receptor FXR and the bile acid excretion pump BSEP (Abcb11) were found to be downregulated, whereas BAAT and Akr1c14, involved in primary bile acid synthesis and bile acid conjugation, were found to be upregulated at mRNA level in GS-positive TSOD mice-derived tumors. BAAT and Akr1c14 were also overexpressed at protein levels. Total cholic acid was found to be increased in GS-positive TSOD mice-derived tumors. CONCLUSION: Our results strongly support the significance of TSOD mice as a model of spontaneously developing HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics , Acyltransferases/genetics , Acyltransferases/metabolism , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Animals , Betaine-Aldehyde Dehydrogenase/genetics , Betaine-Aldehyde Dehydrogenase/metabolism , Cholic Acid/metabolism , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Down-Regulation , Gene Expression , Glutamate-Ammonia Ligase/metabolism , Humans , Male , Mice , Mice, Obese , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Taurocholic Acid/metabolism
6.
Kyobu Geka ; 71(2): 115-119, 2018 Feb.
Article in Japanese | MEDLINE | ID: mdl-29483465

ABSTRACT

We present herein a patient with rocuronium anaphylaxis, which had been identified using skin test, underwent conventional coronary artery bypass surgery without any neuromuscular blocking agent. Immobility was achieved with sedatives and analgesics.


Subject(s)
Anaphylaxis/chemically induced , Coronary Artery Bypass , Coronary Artery Disease/surgery , Neuromuscular Blocking Agents/adverse effects , Aged , Computed Tomography Angiography , Coronary Artery Disease/diagnostic imaging , Humans , Male
7.
Metab Syndr Relat Disord ; 15(4): 170-177, 2017 05.
Article in English | MEDLINE | ID: mdl-28358620

ABSTRACT

BACKGROUND: Metabolic syndrome is one of the most important health issues worldwide. Obesity causes insulin resistance, hyperlipidemia, diabetes, and various diseases throughout the body. The liver phenotype, which is called nonalcoholic steatohepatitis (NASH), frequently progresses to hepatocellular carcinoma. We recently established a new animal model, Tsumura-Suzuki obese diabetic (TSOD) mice, which spontaneously exhibit obesity, diabetes, hyperlipidemia, and NASH with liver nodules. METHODS: We examined the effects of coffee intake on various conditions of the metabolic syndrome using TSOD mice. The daily volume of coffee administered was limited so that it reflected the appropriate quantities consumed in humans. To clarify the effects of the specific components, animals were divided into two coffee-intake groups that included with and without caffeine. RESULTS: Coffee intake did not significantly affect obesity and hyperlipidemia in TSOD mice. In contrast, coffee intake caused various degrees of improvement in the pancreatic beta cell damage and steatohepatitis with liver carcinogenesis. Most of the effects were believed to be caused by a synergistic effect of caffeine with other components such as polyphenols. However, the antifibrotic effects of coffee appeared to be due to the polyphenols rather than the caffeine. CONCLUSIONS: A daily habit of drinking coffee could possibly play a role in the prevention of metabolic syndrome.


Subject(s)
Coffee , Insulin-Secreting Cells/drug effects , Metabolic Syndrome/drug therapy , Non-alcoholic Fatty Liver Disease/prevention & control , Aging , Animals , Caffeine/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/prevention & control , Hyperlipidemias/blood , Insulin-Secreting Cells/pathology , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Liver/pathology , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Liver Neoplasms/epidemiology , Liver Neoplasms/prevention & control , Male , Metabolic Syndrome/complications , Mice , Mice, Obese , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Phosphodiesterase Inhibitors/therapeutic use
8.
Mol Clin Oncol ; 5(2): 267-270, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27446562

ABSTRACT

We previously reported that Tsumura-Suzuki obese diabetic (TSOD) mice, a polygenic model of spontaneous type 2 diabetes, is a valuable model of hepatic carcinogenesis via non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). One of the characteristics of tumors in these mice is the diffuse expression of glutamine synthetase (GS), which is a diagnostic marker for hepatocellular carcinoma (HCC). In this study, we performed detailed histopathological examinations and found that GS expression was diffusely positive in >70% of the hepatic tumors from 15-month-old male TSOD mice. Translocation of ß-catenin into nuclei with enhanced membranous expression also occurred in GS-positive tumors. Small lesions (<1 mm) in GS-positive cases exhibited dysplastic nodules, with severe nuclear atypia, whereas large lesions (>3 mm) bore the characteristics of human HCC, exhibiting nuclear and structural atypia with invasive growth. By contrast, the majority of GS-negative tumors were hepatocellular adenomas with advanced fatty change and low nuclear grade. In GS-negative tumors, loss of liver fatty acid-binding protein expression was observed. These results suggest that the histological characteristics of GS-positive hepatic tumors in TSOD mice resemble human HCC; thus, this model may be a useful tool in translational research targeting the NAFLD/NASH-HCC sequence.

9.
J Toxicol Pathol ; 28(2): 99-107, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26028819

ABSTRACT

A 90-day oral toxicity test in rats was performed to evaluate the toxicity of 2-tetradecylcyclobutanone (2-tDCB), a unique radiolytic product of stearic acid. Six-week-old male and female F344 rats (n=15/group) were given 2-tDCB at concentrations of 0, 12, 60 and 300 ppm in a powder diet for 13 weeks. Slight dose-dependent increases in serum total protein and albumin in male rats were found, but these changes were not considered to be a toxic effect. The fasting, but not non-fasting, blood glucose levels of the male rats in the 300 ppm group and female rats in the 60 and 300 ppm groups were lower than those of the controls. Gas chromatography-mass spectrometry analysis showed dose-dependent accumulation of 2-tDCB in adipose tissue, notably in males. Next, we performed an azoxymethane (AOM)-induced two-stage carcinogenesis study. After injection of 6-week-old male F344 rats (n=30/group) once a week for 3 weeks, the animals received 2-tDCB at concentrations of 0, 10, 50 and 250 ppm in a powder diet for 25 weeks. The incidences of colon tumors for the 2-tDCB dosages were 34%, 45%, 40% and 37%, respectively, and were not statistically significant. These data suggest that 2-tDCB shows no toxic or tumor-modifying effects under the present conditions, and that the no-observed-adverse-effect level for 2-tDCB is 300 ppm in both sexes, equivalent to 15.5 mg/kg b.w./day in males and 16.5 mg/kg b.w./day in females.

10.
Oncotarget ; 6(7): 4829-44, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25638159

ABSTRACT

Associations between growth factor receptor-mediated cell signaling and cancer cell growth have been previously characterized. Receptors for prostaglandin E2, such as EP2, and EP4, play roles in cancer growth, progression and invasion. Thus, we examined the interactions between EP2/EP4- and IGF-1R-mediated cellular signaling in human pancreatic cancer cells. Selective antagonists against EP2 and EP4 abrogated IGF-1-stimulated cell growth and suppressed MEK/ERK phosphorylation. In subsequent experiments, phospho-antibody arrays indicated increased phosphorylation levels of protein kinase C-θ (PKC-θ) at the Thr538 position following the inhibition of EP2/EP4-mediated signaling. Inhibition of PKC-θ activity impaired cell viability compared with EP2/EP4-antagonized IGF-1-stimulated cells. PKC-θ kinase MAP4K3, which plays a pivotal role in PKC-θ activation, also affected growth signaling in the presence of EP2/EP4 antagonists. Administration of EP2 and EP4 antagonists significantly inhibited the growth of an orthotopic xenograft of IGF-1-secreting pancreatic cancer cells, with increased phospho-PKC-θ and decreased phospho-ERK. Clinico-pathological analyses showed that 17.4% of surgical pancreatic cancer specimens were quadruple-positive for IGF-1R, EP2 (or EP4), MAP4K3, and PKC-θ. These results indicate a novel signaling crosstalk between EP2/EP4 and IGF-1R in cancer cells, and suggest that the MAP4K3-PKC-θ axis is central and could be exploited as a molecular target for cancer therapy.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Protein Kinase C-delta/metabolism , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Interactions , Enzyme Activation , Humans , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/metabolism , Isoindoles/pharmacology , Mice , Mice, Nude , Receptor Cross-Talk/drug effects , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Sulfonamides/pharmacology , Xanthones/pharmacology , Xenograft Model Antitumor Assays
11.
Int J Cancer ; 135(11): 2558-68, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-24740818

ABSTRACT

Epidemiologic studies have found that obesity is associated with malignant grade and mortality in prostate cancer. Several adipokines have been implicated as putative mediating factors between obesity and prostate cancer. Fatty acid binding protein 4 (FABP4), a member of the cytoplasmic fatty acid binding protein multigene family, was recently identified as a novel adipokine. Although FABP4 is released from adipocytes and mean circulating concentrations of FABP4 are linked with obesity, effects of exogenous FABP4 on prostate cancer progression are unclear. In this study, we examined the effects of exogenous FABP4 on human prostate cancer cell progression. FABP4 treatment promoted serum-induced prostate cancer cell invasion in vitro. Furthermore, oleic acid promoted prostate cancer cell invasion only if FABP4 was present in the medium. These promoting effects were reduced by FABP4 inhibitor, which inhibits FABP4 binding to fatty acids. Immunostaining for FABP4 showed that exogenous FABP4 was taken up into DU145 cells in three-dimensional culture. In mice, treatment with FABP4 inhibitor reduced the subcutaneous growth and lung metastasis of prostate cancer cells. Immunohistochemical analysis showed that the number of apoptotic cells, positive for cleaved caspase-3 and cleaved PARP, was increased in subcutaneous tumors of FABP4 inhibitor-treated mice, as compared with control mice. These results suggest that exogenous FABP4 might promote human prostate cancer cell progression by binding with fatty acids. Additionally, exogenous FABP4 activated the PI3K/Akt pathway, independently of binding to fatty acids. Thus, FABP4 might be a key molecule to understand the mechanisms underlying the obesity-prostate cancer progression link.


Subject(s)
Cell Movement , Cell Proliferation , Fatty Acid-Binding Proteins/metabolism , Lung Neoplasms/secondary , Prostatic Neoplasms/pathology , Animals , Apoptosis , Blotting, Western , Disease Progression , Fatty Acids/metabolism , Flow Cytometry , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Int J Oncol ; 43(2): 365-74, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23708710

ABSTRACT

New drugs that inhibit the osteoprotegerin (OPG)/receptor activator of NF-κB ligand (RANKL)/RANK pathway have demonstrated efficacy for the treatment of bone metastasis. Toxicities induced by these drugs, however, including osteonecrosis of the jaw and hypocalcemia, may adversely affect therapy. The aim of this study was to identify additional therapeutic targets that can be combined with OPG/RANKL/RANK pathway inhibition in the treatment of prostate cancer bone metastasis. We established a stable transfectant that produces high levels of OPG mRNA and protein from PC-3 human prostate cancer cells (PC3-OPG). The culture medium of PC3-OPG cells significantly inhibited the differentiation of mouse monocytes into mature osteoclasts. Furthermore, when PC3-OPG cells were injected into the bones of nude mice, bone destruction and tumor-induced osteoclast formation were reduced. Injection into bone of the mixtures containing equal amounts of green fluorescent protein (GFP)-expressing PC-3 cells (PC3-GFP) and PC3-OPG cells also reduced bone destruction, compared to the control mixture. PC3-GFP cells were subsequently isolated from bone tumors and used for microarray analysis to assess changes in gene expression following osteolytic tumor growth inhibition by OPG. We selected the top 10 upregulated genes based on results from microarrays and confirmed mRNA expression of each gene by RT-PCR. The expression patterns of retinol-binding protein 4 (RBP4) and placenta-specific 8 (PLAC8) were consistent with microarray results. Expression of these genes was also increased in the bone tumors of PC3-GFP/PC3-OPG-injected mice. Knockdown of both RBP4 and PLAC8 by siRNA inhibited the growth of PC-3 cells in vitro. Thus, RBP4 and PLAC8 may become new therapeutic targets for prostate cancer bone metastasis, in combination with OPG/RANKL/RANK pathway inhibition.


Subject(s)
Osteolysis, Essential/metabolism , Osteoprotegerin/metabolism , Prostatic Neoplasms/metabolism , Proteins/metabolism , Retinol-Binding Proteins, Plasma/metabolism , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Green Fluorescent Proteins/genetics , Humans , Male , Mice , Mice, Nude , Osteoclasts/metabolism , Osteolysis, Essential/genetics , Osteoprotegerin/biosynthesis , Osteoprotegerin/genetics , Prostatic Neoplasms/genetics , Proteins/genetics , RANK Ligand/metabolism , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Receptor Activator of Nuclear Factor-kappa B/metabolism , Retinol-Binding Proteins, Plasma/biosynthesis , Retinol-Binding Proteins, Plasma/genetics
13.
Int J Oncol ; 43(2): 416-24, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23708855

ABSTRACT

We have previously reported that FuEP2/Ex2, a soluble decoy receptor for PGE2, suppresses tumor growth in an orthotopic xenograft model. To examine whether it has further uses, we examined the effect of FuEP2/Ex2 in an intraperitoneal metastasis model of ovarian cancer cells. We established FuEP2/Ex2-expressing ovarian cancer cells (SKOV/ip-FuEP2/Ex2) and injected them intraperitoneally into female nude mice. Mice injected with SKOV/ip-FuEP2/Ex2 had no ascitic fluid and showed smaller tumor lesions compared to mice injected with vector control cells, with decreased microvessel density and M2 macrophages. To identify molecular targets for combination treatment, we conducted cDNA microarray analysis and found three genes encoding enzyme [matrix metalloproteinase-7 (MMP-7), transmembrane protease serin 4 (TMPRSS4) and cytocrome P450 1B1 (CYP1B1)] to be upregulated in SKOV/ip-FuEP2/Ex2-derived tumors. Administration of TMPRSS4 inhibitor further reduced tumor weight and decreased the number of Ki-67­positive cells in SKOV/ip-FuEP2/Ex2-injected mice. These data indicate a possible EP-targeting strategy using FuEP2/Ex2 in the treatment of ovarian cancer and suggest that dual targeting of EP-mediated signaling and TMPRSS4 may enhance therapeutic value.


Subject(s)
Membrane Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Serine Endopeptidases/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/biosynthesis , Aryl Hydrocarbon Hydroxylases/genetics , Ascitic Fluid , Cell Proliferation/drug effects , Chemokine CXCL1/metabolism , Cytochrome P-450 CYP1B1 , Female , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Ki-67 Antigen/metabolism , Matrix Metalloproteinase 7/biosynthesis , Matrix Metalloproteinase 7/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Ovarian Neoplasms/genetics , RNA Interference , RNA, Small Interfering , Receptors, Prostaglandin E, EP2 Subtype/genetics , Serine Endopeptidases/genetics , Serine Proteinase Inhibitors/pharmacology , Signal Transduction , Sulfones/pharmacology , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
14.
Pathol Int ; 62(6): 418-23, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22612511

ABSTRACT

Balamuthia mandrillaris is a free-living ameba that causes amebic encephalitis. Herein, we report an autopsy case of Balamuthia encephalitis proven with polymerase chain reaction (PCR) and immunohistochemistry from paraffin-embedded brain biopsy specimens. A 68-year-old Japanese male presented at a hospital with progressive right hemiparesis approximately 3 months before his death. An open-brain biopsy specimen showed diffuse meningitis with massive coagulative necrosis. The perivascular spaces contained numerous lymphocytes, histiocytes and giant cells, although the etiology was not determined. The patient deteriorated into coma and died from cerebral herniation. Autopsy revealed abundant trophozoites and cysts in the subarachnoid and Virchow-Robin's spaces. Electron-micrographs of the amebic cysts showed a characteristic triple-walled envelope. The amebas were identified as Balamuthia mandrillaris based on immunohistochemical analysis from the autopsy and biopsy specimens. Primer sets designed to amplify approximately 200 bp bands of mitochondrial 16S rRNA gene of Balamuthia by PCR produced positive results from the biopsy specimens but negative results from the autopsy specimens. In summary, PCR to amplify shorter segments of DNA may be of diagnostic value in detecting suspected cases of balamuthiasis in formalin-fixed, paraffin-embedded specimens. Increased awareness and timely diagnosis of Balamuthia encephalitis might lead to earlier initiation of therapy and improved outcome.


Subject(s)
Amebiasis/pathology , Balamuthia mandrillaris/isolation & purification , Brain/pathology , Central Nervous System Protozoal Infections/pathology , Amebiasis/drug therapy , Amebiasis/parasitology , Balamuthia mandrillaris/physiology , Brain/parasitology , Central Nervous System Protozoal Infections/drug therapy , Central Nervous System Protozoal Infections/parasitology , Fatal Outcome , Humans , Japan , Magnetic Resonance Imaging , Male , Treatment Failure
15.
Cancer Sci ; 102(10): 1904-10, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21733044

ABSTRACT

Bone metastasis is a frequent complication of advanced breast cancer. On the basis of functional and molecular evidence, signaling mediated by the binding of platelet-derived growth factor (PDGF)-BB and -DD to PDGF receptor ß (PDGFRß) is critical for the survival and growth of metastatic breast cancer cells within the bone microenvironment. In this study, we propose a new approach to blocking PDGFRß signaling using soluble PDGFRß (sPDGFRß) as a decoy receptor for PDGF-BB and -DD secreted from tumor cells and bone marrow stromal cells. A bone-seeking TNBCT/Bo cell line was established by in vivo selection from TNBCT human breast cancer cells, which are negative for estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 protein expression. The TNBCT/Bo cells were transfected with a mammalian expression vector encoding the extracellular domain of PDGFRß. A stable transfectant (TNBCT/Bo-sPDGFRß) grew at a similar rate to that of control cells under normal culture conditions, although growth stimulation of human fibroblasts with PDGF-BB was neutralized by the culture medium from TNBCT/Bo-sPDGFRß cells. Intratibial injection of TNBCT/Bo-sPDGFRß cells into athymic nude mice resulted in a significant decrease in tumor incidence compared with control mice (P < 0.01). This attenuated growth correlated with decreased cancer cell proliferation, angiogenesis, and recruitment of stromal cells, and with an increase in the number of apoptotic cells. These findings suggest that sPDGFRß is useful for the treatment of breast cancer bone metastasis.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Receptor, Platelet-Derived Growth Factor beta/pharmacology , Animals , Apoptosis , Becaplermin , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/biosynthesis , Female , Gene Transfer Techniques , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/biosynthesis , Signal Transduction/drug effects , Solubility , Stromal Cells/drug effects , Stromal Cells/metabolism
16.
Cancer Lett ; 306(1): 67-75, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21419570

ABSTRACT

Endometrial cancer is one of the most common gynecologic malignancies and many factors influence in its growth and development. As in many other types of cancer, prostaglandin E(2) (PGE(2)) is thought to be an accelerator of cell proliferation and endometrial cancer progression. In this study, we examined the effect of FuEP2/Ex2, a soluble decoy receptor for PGE(2) on growth of endometrial cancer cells. A stable transfectant expressing FuEP2/Ex2 was established from human endometrial cancer Ishikawa cells (Ish-FuEP2/Ex2). Ish-FuEP2/Ex2 cells expressed FuEP2/Ex2 mRNA and protein. Expression levels of E-prostanoid receptor 1 (EP1), EP2, EP3, EP4, and F-prostanoid receptor (FP) were almost the same in Ish-FuEP2/Ex2 and vector control cells. Growth rates of Ish-FuEP2/Ex2 under normal culture conditions were also similar to vector control cells, although PGE(2)-induced growth stimulation was completely inhibited in Ish-FuEP2/Ex2 or by Ish-FuEP2/Ex2 culture medium. Moreover, phosphorylation of extracellular signal-regulated kinase (ERK) and induction of cyclooxygenase-2 (COX-2), vascular endothelial growth factor (VEGF), cyclin D1, and c-fos mRNA by PGE(2) were not observed in Ish-FuEP2/Ex2 and Ish-FuEP2/Ex2 culture medium-treated vector control cells, although they were found when treated with prostaglandin F(2α). An orthotopic xenograft model in athymic nude mice revealed that Ish-FuEP2/Ex2-injected mice had significantly decreased mean tumor area. The proportion of Ki-67-positive cells in the tumor lesion was also significantly lower in Ish-FuEP2/Ex2-injected mice. These findings suggest that an EP-targeting strategy using FuEP2/Ex2 may be of use in the treatment of endometrial cancer.


Subject(s)
Endometrial Neoplasms/metabolism , Receptors, Prostaglandin E, EP2 Subtype/chemistry , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transplantation , Culture Media , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Female , Humans , Ki-67 Antigen/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Receptors, Prostaglandin E, EP2 Subtype/metabolism
17.
J Toxicol Pathol ; 24(4): 187-93, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22319230

ABSTRACT

Diabetes mellitus (DM) and obesity are believed to be risk factors for colorectal cancer in humans. In experiment 1, male nondiabetic Long-Evans Tokushima Otsuka (LETO) rats and Otsuka Long-Evans Tokushima Fatty (OLETF) rats, a model animal of type 2 DM, were whole-body X-irradiated (4 Gy) at 6 and 8 weeks of age and euthanized at 78 weeks of age (n=15, respectively). The incidences of small intestine adenocarcinoma in LETO and OLETF rats were 0% and 30%, respectively. In experiment 2, male LETO and OLETF rats (n=24, respectively) were given s.c. injections of 15 mg/kg azoxymethane (AOM) once weekly for 3 weeks and euthanized at 36 weeks of age. The incidences of Zymbal gland tumors in LETO and OLETF rats were 0% and 67%, respectively (P<0.001), whereas those of small intestine adenocarcinoma were 0% and 43% (P<0.001) and those of cecum/colon adenocarcinoma were 46% and 79% (P<0.05), respectively. Fatty change of hepatocytes was common in OLETF rats (63%) but not in LETO rats. Serum triglyceride and free fatty acid levels in OLETF rats were significantly higher than in LETO rats at sacrifice, whereas serum insulin levels in OLETF rats were very diverse. These data suggest that hyperlipidemia plays a significant role in high susceptibility to lower intestinal tract carcinogenesis in OLETF rats; this strain is susceptible to AOM-induced Zymbal gland carcinogenesis.

18.
Masui ; 59(2): 179-82, 2010 Feb.
Article in Japanese | MEDLINE | ID: mdl-20169953

ABSTRACT

BACKGROUND: Transient renal failure during surgery is caused by increasing secretion of stress hormone such as ADH and renin. We suspected that urinary output varies according to administration of remifentanil with potent analgesic effects. Consequently, we studied intraoperative urinary output of two groups, patients administered with remifentanil and those without remifentanil administration. METHODS: We compared urinary output during general anesthesia, of 327 patients administered with remifentanil (Group R) and 314 patients without remifentanil administration (Group NR) retrospectively. Patients were excluded if they were under the age of eighteen, receiving epidural anesthesia, or having medicine with diuretic effect. RESULTS: There were no significant difference in background of the patients in each group, in particular, age, sex, body weight, and ASA grade. We found no significant difference in intraoperative factors; operation time, total blood loss, volume of infusion, anesthesia time, and given dose of fentanyl. Urinary output of Group R was estimated as 512 +/- 435 ml, and that of Group NR was 409 +/- 405 ml (P value was 0.02). CONCLUSIONS: We found a significance difference in urinary output during anesthesia, between patients administered with remifentanil and those without remifentanil administration. We suspect that remifentanil decreases urinary output in the perioperative period.


Subject(s)
Analgesics, Opioid/pharmacology , Anesthesia, General , Piperidines/pharmacology , Urination/drug effects , Adult , Aged , Analgesics, Opioid/administration & dosage , Female , Humans , Intraoperative Period , Male , Middle Aged , Piperidines/administration & dosage , Remifentanil , Retrospective Studies
19.
Oncol Lett ; 1(6): 981-987, 2010 Nov.
Article in English | MEDLINE | ID: mdl-22870098

ABSTRACT

Nucleic acid-metabolizing enzymes, such as thymidylate synthase (TS), dihydropyrimidine dehydrogenase (DPD), thymidine phosphorylase (TP) and orotate phosphoribosyl transferase (OPRT), have attracted attention as candidates for response determinants of 5-fluorouracil (5-FU). Whether the expression levels of these enzymes can be adopted as valuable parameters for 5-FU sensitivity in breast cancer has yet to be elucidated. In the present study, intratumoral mRNA expression of TS, DPD, TP and OPRT were determined in formalin-fixed paraffin-embedded surgical specimens collected from 217 breast cancer patients, using the Danenberg Tumor Profile method, which combines microdissection and real-time-polymerase chain reaction. The significance of these enzymes as prognostic and 5-FU efficacy-predicting factors was evaluated. Our data showed that a low DPD expression is related to a high nuclear grade and other factors including hormone receptor-negativity. Low expression levels of TP were found in hormone receptor-negative tumors. TS and OPRT expression were not related to various clinicopathological factors, but patients with a high TS mRNA expression showed a significantly poorer prognosis in cases where 5-FU was not administered. The efficacy of 5-FU was more significant when administered for more than 6 months in the group with a high TS mRNA expression. These data suggest that TS mRNA expression in breast cancer tissue is an ideal predictor of outcomes for patients with no administration of 5-FU, and of the efficacy of 5-FU.

20.
J Med Invest ; 56(3-4): 93-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19763020

ABSTRACT

The Long-Evans Cinnamon (LEC) rat, an animal model of Wilson's disease, is resistant to a variety of chemical carcinogenesis except liver and colon. In the present study, N-ethyl-N-nitrosourea (ENU)-induced transplacental carcinogenesis was examined in male and female LEC, Long-Evans Agouti (LEA), a sibling line of the LEC rat, and F344 rats (n=21). ENU was administered to pregnant rats as a single s.c. injection at a dose of 60 mg/kg body weight on the 17th day after conception. Cerebral/spinal gliomas and trigeminal/spinal nerve schwannomas developed in both LEA and F344 rats at 30 weeks of age, but no nervous system tumors developed in LEC rats, the difference being statistically significant. Lung adenomas also developed in LEA and F344 rats, but not in LEC rats. Semiquantitative RT-PCR demonstrated that metallothionein (MT)1a, MT2 and O(6)-methylguanine-DNA methyltransferase (MGMT) mRNA levels in the liver of LEC rats were higher than those in F344 and LEA rats. In addition, Western blot analysis showed that MT (MT1 plus MT2) in the liver of LEC rats was also higher than that in other strains. Present results suggest that high levels of MT and/or MGMT contribute to the resistance to nitrosamine-induced carcinogenesis in LEC rats.


Subject(s)
Carcinogens/toxicity , Ethylnitrosourea/toxicity , Maternal-Fetal Exchange , Animals , Base Sequence , Carcinogens/administration & dosage , Ethylnitrosourea/administration & dosage , Female , Liver/drug effects , Liver/metabolism , Lung Neoplasms/chemically induced , Male , Metallothionein/genetics , Nervous System Neoplasms/chemically induced , O(6)-Methylguanine-DNA Methyltransferase/genetics , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Inbred LEC , Rats, Long-Evans , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...