Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 22(12): 1454-1464, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37683276

ABSTRACT

Topotecan, an approved treatment for refractory or recurrent ovarian cancer, has clinical limitations such as rapid clearance and hematologic toxicity. To overcome these limitations and maximize clinical benefit, we designed FF-10850, a dihydrosphingomyelin-based liposomal topotecan. FF-10850 demonstrated superior antitumor activity to topotecan in ovarian cancer cell line-based xenograft models, as well as in a clinically relevant DF181 platinum-refractory ovarian cancer patient-derived xenograft model. The safety profile was also improved with mitigation of hematologic toxicity. The improved antitumor activity and safety profile are achieved via its preferential accumulation and payload release triggered in the tumor microenvironment. Our data indicate that tumor-associated macrophages internalize FF-10850, resulting in complete payload release. The release mechanism also appears to be mediated by high ammonia concentration resulting from glutaminolysis, which is activated by tumor metabolic reprogramming. In ammonia-rich conditions, FF-10850 released payload more rapidly and to a greater extent than liposomal doxorubicin, a currently approved treatment for ovarian cancer. FF-10850 significantly enhanced antitumor activity in combination with carboplatin or PARP inhibitor without detrimental effects on body weight in murine xenograft models, and demonstrated synergistic antitumor activity combined with anti-PD-1 antibody with the development of tumor antigen-specific immunity. These results support phase I investigation of FF-10850 for the treatment of solid tumors including ovarian cancer (NCT04047251), and further evaluation in combination settings.


Subject(s)
Ovarian Neoplasms , Topotecan , Female , Humans , Animals , Mice , Topotecan/pharmacology , Ammonia/therapeutic use , Tumor Microenvironment , Ovarian Neoplasms/pathology , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/therapeutic use , Macrophages/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
2.
J Cancer Res Clin Oncol ; 139(3): 395-401, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23114882

ABSTRACT

PURPOSE: Some goitrogens promote thyroid carcinogenesis in rats in an initiation-promotion model; this model frequently produces carcinomas that invade fibrously thickened capsules, termed capsular invasive carcinomas (CICs). The present study tested a hypothesis that CICs originate from parenchymal proliferative lesions located beneath the capsule. METHODS: Cell proliferation activity, cell-cycle kinetics and cellular invasion were immunohistochemically examined in subcapsular proliferative lesions in male F344 rats treated with an anti-thyroid agent, propylthiouracil or sulfadimethoxine, during the tumor-promotion phase after initiation with N-bis(2-hydroxypropyl)nitrosamine. RESULTS: Focal follicular cell hyperplasias (FFCHs) were the most commonly observed parenchymal proliferative lesions. Subcapsular FFCHs located near CICs showed more Ki-67(+) cells in the capsular side than the contrary parenchymal center side. Most of these FFCHs located near CICs showed accumulated immunoreactivity for cyclin A, cyclin D, cyclin E and cyclin-dependent kinase-2, whereas most subcapsular FFCHs located elsewhere did not show such accumulated expression of cell-cycle molecules. Subcapsular FFCHs immunoreactive at the capsular front for tenascin-C, a tumor invasion marker of extracellular matrix protein, showed high proliferation activity. CONCLUSIONS: Subcapsular FFCH-forming cells can potentially spread directly into the fibrously thickened capsule to form CICs by accelerating cell-cycle activity.


Subject(s)
Carcinoma/pathology , Disease Models, Animal , Hypothyroidism/complications , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Animals , Carcinogens , Carcinoma/chemically induced , Carcinoma/etiology , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/pathology , Disease Progression , Hyperplasia/etiology , Hyperplasia/pathology , Hypothyroidism/pathology , Male , Neoplasm Invasiveness , Nitrosamines , Rats , Rats, Inbred F344 , Thyroid Neoplasms/chemically induced , Thyroid Neoplasms/etiology
3.
Transl Oncol ; 6(6): 628-37, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24466365

ABSTRACT

This study evaluated the detection of tumors using in vivo imaging with a commercially available and systemically administered protease-activatable fluorescent probe, ProSense. To this end, we analyzed the delivery and uptake of ProSense as well as the target protease and its cellular source in a mouse xenograft tumor model. In vivo and ex vivo multi wavelength imaging revealed that ProSense signals accumulated within tumors, with preferential distribution in the vascular leakage area that correlates with vasculature development at the tumor periphery. Immunohistochemically, cathepsin B, which is targeted by ProSense, was specifically localized in macrophages. The codistribution of tenascin C immunoreactivity and gelatinase activity provided evidence of tissue-remodeling at the tumor periphery. Furthermore, in situ zymography revealed extracellular ProSense cleavage in such areas. Colocalization of cathepsin B expression and ProSense signals showing reduction by addition of cathepsin B inhibitor was confirmed in cultured macrophage-derived RAW264.7 cells. These results suggest that increased tissue-remodeling activity involving infiltration of macrophages is a mechanism that may be responsible for the tumor accumulation of ProSense signals in our xenograft model. We further confirmed ProSense signals at the tumor margin showing cathepsin B(+) macrophage infiltration in a rat colon carcinogenesis model. Together, these data demonstrate that systemically administered protease-activatable probes can effectively detect cancer invasive fronts, where tissue-remodeling activity is high to facilitate neoplastic cell invasion.

4.
J Toxicol Pathol ; 25(2): 149-54, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22907981

ABSTRACT

Activators of tissue proteolysis including Stachybotrys microspora triprenyl phenol (SMTP)-7 are a new class of agents that are expected to be effective for amelioration of chronic tissue destructive diseases. The present study was performed to examine whether SMTP-7 is effective for the amelioration or protection of early-stage IgA nephropathy (IgAN) induced by nivalenol (NIV) in female BALB/c mice. In Experiment 1, mice were administered NIV at 24 ppm in diet for 8 weeks, and during the NIV treatment, they were intraperitoneally injected with SMTP-7 (10 mg/kg) three times a week. In Experiment 2, mice were injected similarly with SMTP-7 during the last 4 weeks of a 16-week NIV treatment. Immunofluorescence analysis revealed an inhibitory effect of SMTP-7 on the glomerular deposition of IgA in Experiment 1; however, it was ineffective in Experiment 2. On the other hand, SMTP-7 did not affect the serum concentration of IgA in both experiments. These results suggest that SMTP-7 has a potential to decrease the progression of IgAN induced by NIV through inhibition of local accumulation of IgA in the glomerular mesangium, while it was ineffective for suppression of IgA production. On the other hand, SMTP-7 was found to be ineffective for already deposited IgA, suggesting that SMTP-7 may not be effective for ameliorating advanced IgAN.

5.
J Toxicol Sci ; 37(3): 475-82, 2012.
Article in English | MEDLINE | ID: mdl-22687987

ABSTRACT

Cochineal extracts (CE) is a coccid-derived natural food colorant containing carminic acid (CA) as an active ingredient that potentiates inhibition of tissue proteolysis mediated by activation of plasma hyaluronan-binding protein (PHBP). In our previous study, dietary administered CE (CA: 28.5% in CE) has shown to promote the macroscopic development of capsular invasive carcinomas (CICs) associated with up-regulation of angiogenesis-related genes in an intracapsular invasion model of experimental thyroid cancers using rats. However, the promoting effect of CE could not be confirmed histopathologically. The purpose of the present study was to confirm the promoting effect of CE through direct injections to animals on the development of CICs using this cancer invasion model. One week after initiation with N-bis(hydroxypropyl)nitrosamine, male F344/NSlc rats were administered CA-enriched CE (CA: 52.6% in CE) by intraperitoneal injections every other day (10 mg/kg body weight) during the promotion with 0.15% sulfadimethoxine in the drinking water for 8 weeks. The multiplicities of macroscopical CICs and the mean area of early capsular invasive foci estimated by Tenascin (TN)-C-immunoreactivity in the thyroid significantly increased with CE-treatment, while the number of TN-C-positive foci did not change with CE. Transcript level of Phbp and downstream genes unchanged; however, transcript level of angiogenesis-related genes, i.e, Vegfb and its transcription factor gene, Hif1a, those being downstream of phosphatase and tensin homolog (PTEN)/Akt signaling, up-regulated in the thyroid tissue with CE-administration. These results suggest that CE potentiates promotion activity by facilitating angiogenesis through activation of PTEN/Akt signaling without accompanying modification of PHBP-related proteolysis.


Subject(s)
Carmine/analogs & derivatives , Plant Extracts/pharmacology , Thyroid Neoplasms/drug therapy , Angiogenesis Inducing Agents/therapeutic use , Animals , Carmine/pharmacology , Disease Models, Animal , Drinking Water/administration & dosage , Drinking Water/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Nitrosamines/toxicity , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Inbred F344 , Signal Transduction , Sulfadimethoxine/metabolism , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroid Gland/pathology , Thyroid Neoplasms/chemically induced , Thyroid Neoplasms/pathology , Up-Regulation
6.
Exp Biol Med (Maywood) ; 237(6): 728-38, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22715430

ABSTRACT

Coccid-derived natural food colorants contain active ingredients that potentiate inhibition of tissue proteolysis mediated by activation of plasma hyaluronan-binding protein (PHBP). In the present study, we examined the effect of lac color (LC) and cochineal extract (CE), representative coccid-derived colorants containing laccaic acid and carminic acid as active ingredients, in an intracapsular invasion model of experimental thyroid cancers using rats. One week after initiation with N-bis(hydroxypropyl)nitrosamine, male F344/NSIc rats were fed a powdered diet containing 5.0% LC or 3.0% CE during promotion with 0.15% sulfadimethoxine (SDM) in the drinking water for 13 weeks. Capsular invasive carcinomas (CICs) and lung metastases were decreased by LC treatment and accompanied by transcript downregulation on angiogenesis and PHBP-related tissue proteolysis in CICs. In contrast, CE upregulated angiogenesis-related genes in CICs. PHBP was expressed in capsular macrophages and thyroid proliferative lesions with increased intensity in CICs, and LC decreased PHBP-expressing CICs. The size of CICs and their proliferation activity, however, were unchanged compared with those treated with SDM alone. Suppression of cancer by invasion by LC was more evident after an eight-week treatment, exhibiting a profound decrease in tenascin-C-positive early invasive foci and marked reductions in capsular inflammation and fibrosis. These results suggest that LC and CE exerted dissimilar effects on CIC development, the former suppressing the initial step of neoplastic cell invasion into the capsule by targeting PHBP activity of macrophages and neoplastic cells on tissue proteolysis involving inflammatory responses and angiogenesis, and the latter promoting angiogenesis of developed CICs at later stages.


Subject(s)
Azo Compounds/therapeutic use , Carcinoma, Papillary, Follicular/drug therapy , Carcinoma, Papillary, Follicular/physiopathology , Hyaluronan Receptors/blood , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/physiopathology , Animals , Carcinoma, Papillary, Follicular/chemically induced , Cell Proliferation , Disease Models, Animal , Male , Naphthalenesulfonates , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Inbred F344 , Signal Transduction/physiology , Sulfadimethoxine/adverse effects , Thyroid Hormones/blood , Thyroid Neoplasms/chemically induced
7.
Toxicology ; 283(2-3): 109-17, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21396975

ABSTRACT

The liver tumor-promoting effects of indole-3-carbinol (I3C), a cytochrome P450 (CYP) 1A inducer found in cruciferous vegetables, were investigated using a medium-term hepatocarcinogenesis model in rats. Six-week-old male F344 rats received an intraperitoneal injection of N-diethylnitrosamine (DEN) and were fed a diet containing 0 (DEN-alone), 0.25, 0.50 or 1.0% of I3C for 8 weeks from 2 weeks after DEN-initiation. The number and area of liver cell foci positive for glutathione S-transferase placental form (GST-P) significantly increased in the livers of rats given 0.5% I3C or more, compared to those in the DEN-alone group. The number of GST-P positive foci also increased in the 0.25% I3C group. The number of liver cells positive for proliferating cell nuclear antigen (PCNA) significantly increased in all I3C groups compared to that in the DEN-alone group. Real-time RT-PCR analysis showed that I3C increased transcript levels of not only Cyp1a1 but also aryl hydrocarbon receptor (AhR) and/or nuclear factor (erythroid-derived 2)-like 2 (Nrf2) gene batteries, such as Cyp1a2, Cyp1b1, Ugt1a6, Nrf2, Nqo1, Gsta5, Gstm2, Ggt1and Gpx2. Reactive oxygen species (ROS) in the microsomal fraction significantly increased in all I3C-treated groups compared to the DEN-alone group, and thiobarbituric acid-reactive substances (TBARS) levels and 8-hydroxy-2'-deoxyguanosine (8-OHdG) content significantly increased in all of the I3C-treated groups and 1.0% I3C group, respectively. These results suggest that I3C is an AhR activator and enhances microsomal ROS production resulting in the upregulation of Nrf2 gene batteries, but the oxidative stress generated overcomes the antioxidant effect of Nrf2-related genes. Such 'a redox imbalance' subsequently induces liver tumor-promoting effects by enhancing cellular proliferation in rats.


Subject(s)
Carcinogens/pharmacology , Diethylnitrosamine/pharmacology , Free Radical Scavengers/pharmacology , Indoles/pharmacology , Liver Neoplasms, Experimental/pathology , Oxidative Stress/drug effects , Precancerous Conditions/chemically induced , 8-Hydroxy-2'-Deoxyguanosine , Animals , Body Weight/drug effects , DNA Damage/drug effects , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Enzyme Induction/drug effects , Hepatectomy , Immunohistochemistry , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Liver Neoplasms, Experimental/metabolism , Male , Microarray Analysis , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Organ Size/drug effects , Precancerous Conditions/pathology , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thiobarbituric Acid Reactive Substances/metabolism
8.
J Vet Med Sci ; 73(5): 655-64, 2011 May.
Article in English | MEDLINE | ID: mdl-21206174

ABSTRACT

We have shown phosphoinositide 3-kinase (PI3K)/Akt signaling activation in thyroid capsular invasive carcinomas (CICs), which are highly induced by promotion with sulfadimethoxine (SDM) in a rat 2-stage thyroid carcinogenesis model. To examine the potency of calcitriol, a synthetic vitamin D3 analog, on the development or progression of CICs, male F344 rats were injected with calcitriol (0.1 µg/kg body weight) three times a week intraperitoneally, during an entire period of SDM-promotion for 13 weeks (Experiment 1) or during the last 2 weeks of a 15-week SDM-promotion (Experiment 2). Initiation with N-bis(2-hydroxypropyl)nitrosamine preceded all treatments. In Experiment 1, long-term calcitriol treatment reduced the multiplicity of CICs, while cell proliferation activity, estimated by Ki-67 cell index in the induced CICs, was unchanged with SDM-promotion alone. Considering the strong dependency of promotion with SDM during the early stages on thyroid-stimulating hormone, the reduced multiplicity in Experiment 1 may be due to the effect on an early stage of neoplastic proliferation. Although the magnitude was mild, cell proliferation activity was decreased in existing CICs after short-term calcitriol treatment in Experiment 2, which was associated with a mild decrease in cyclin-dependent kinase-2-positive cells, cytoplasmic immunolocalization of phosphorylated, inactive, Rb protein and a mild increase in nucleocytoplasmic expression of p27(kip1). Although the effect was mild at the late stage of SDM-promotion in this hypothyroidism-related thyroid carcinogenesis model, our results suggest that calcitriol targets cell proliferation via inhibition of a molecular cascade downstream of PI3K/Akt signaling, controlling G1/S transition.


Subject(s)
Calcitriol/pharmacology , Calcium Channel Agonists/pharmacology , Carcinoma/prevention & control , Thyroid Neoplasms/prevention & control , Animals , Apoptosis/drug effects , Body Weight , Male , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Inbred F344 , Signal Transduction , Sulfadimethoxine/toxicity
9.
Arch Toxicol ; 85(9): 1159-66, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21203749

ABSTRACT

To investigate liver tumor-promoting potentials of indole-3-carbinol (I3C) and flutamide (FLU), changes in mRNA expression of Cyp1a and genes encoding antioxidant/detoxifying enzymes in the liver, 6-week-old male F344 rats were subjected to medium-term liver bioassay. ß-Naphthoflavone (BNF), a strong CYP1A inducer, was also used for comparison. Two weeks after initiation with N-diethylnitrosamine (DEN), animals were fed a basal diet (untreated controls) or a diet containing 0.5% I3C, 0.1% FLU, or 0.5% BNF for 6 weeks. Each animal was subjected to a two-third partial hepatectomy 1 week after the start of promoter treatments. Histopathologically, I3C and BNF increased altered liver cell foci with the incidence (3.7- and 7.3-fold) and multiplicity (8.3- and 13.8-fold) compared with the DEN-alone group, respectively. Immunohistochemically, I3C significantly increased the number (3.1-fold; P < 0.01) and area (2.4-fold; P < 0.05) of foci positive for glutathione-S-transferase placental form (GST-P) compared with the DEN-alone group; FLU induced a slight but significant increase in the number of GST-P-positive foci (2.8-fold; P < 0.05) whereas BNF showed marked induction of the number and area of GST-P-positive foci (20- and 14-fold, respectively; P < 0.01). In parallel, I3C, FLU, and BNF markedly increased mRNA levels of Cyp1a1 (50-, 23-, 299-fold) and antioxidant/detoxifying enzymes such as Gpx2 and Nqo1 as shown by real-time reverse transcription-polymerase chain reaction analysis. These results suggest that I3C and FLU could promote hepatocellular tumors in parallel with that of CYP1A's potential to cause subsequent oxidative stress responses in rats.


Subject(s)
Cytochrome P-450 CYP1A1/biosynthesis , Flutamide/toxicity , Indoles/toxicity , Liver Neoplasms, Experimental/enzymology , Liver/drug effects , Animals , Body Weight/drug effects , Cytochrome P-450 CYP1A1/genetics , Diethylnitrosamine/toxicity , Enzyme Induction , Gene Expression/drug effects , Glutathione S-Transferase pi/metabolism , Hepatectomy , Immunohistochemistry , Liver/enzymology , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Organ Size/drug effects , Oxidative Stress/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , beta-Naphthoflavone/pharmacology
10.
Exp Toxicol Pathol ; 63(1-2): 17-24, 2011 Jan.
Article in English | MEDLINE | ID: mdl-19783131

ABSTRACT

To clarify the underlying mechanisms of IgA nephropathy (IgAN) induced by nivalenol (NIV), a trichothecene mycotoxin, we examined the time and dose relationships of glomerular deposition of IgA by NIV in BALB/c mice (Experiment 1), and also evaluated the modification of NIV on spontaneous IgAN in an inbred murine model, a high IgA strain (HIGA), during its early stage of pathogenesis (Experiment 2). In Experiment 1, female BALB/c mice were given a diet containing 0, 12, or 24 ppm concentration of NIV for 4 or 8 weeks. An increase in serum IgA levels was found at 24 ppm from 4 weeks. At week 8 of treatment, dose-dependent increases in serum IgA levels and glomerular deposition of IgA and IgG were observed without accompanying histopathological glomerular changes. On the other hand, in Experiment 2, control HIGA mice exhibited rather high levels of serum IgA as compared with BALB/c mice from 4 weeks of experiment as well as glomerular deposition of IgA and IgG and mesangial proliferation as revealed at week 8. NIV at 24ppm further increased serum IgA in this strain; however, it did not enhance glomerular immunoglobulin deposition or histopathological lesion. These results suggest that NIV-induced increase of serum IgA levels may be primarily responsible for glomerular immunoglobulin deposition; however, NIV does not enhance glomerular IgA deposition that may lead to exacerbation of predisposed IgAN in the short term, irrespective of the further elevation of serum IgA from the high basal levels.


Subject(s)
Glomerular Mesangium/pathology , Glomerulonephritis, IGA/chemically induced , Immunoglobulin A/blood , Trichothecenes/toxicity , Animals , Dose-Response Relationship, Drug , Female , Fluorescent Antibody Technique, Indirect , Glomerular Mesangium/metabolism , Glomerulonephritis, IGA/blood , Glomerulonephritis, IGA/metabolism , Glomerulonephritis, IGA/pathology , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C
11.
J Cancer Res Clin Oncol ; 137(4): 723-32, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20582435

ABSTRACT

PURPOSE: Rat thyroid follicular cell carcinomas invading into the thyroid capsule are highly produced by promotion with sulfadimethoxine (SDM) in a rat two-stage thyroid carcinogenesis model. In this study, we investigated the participation of phosphoinositide 3-kinase (PI3K) signaling pathway that is associated with malignant phenotypes of many cancers on the development of SDM-induced capsular invasive carcinomas. METHODS: Thyroid proliferative lesions developed 10 or 15 weeks after promotion with SDM in male F344 rats initiated with N-bis(2-hydroxypropyl)nitrosamine were immunohistochemically analyzed with regard to cellular distribution of phosphatase and tensin homolog (PTEN) and Akt isoforms, as well as their downstream molecules. RESULTS: Increased expression of PI3K signaling molecules was evident in association with the development of lesion stages from the early focal hyperplasia to the late carcinomas. Capsular carcinomas, and the less frequent parenchymal carcinomas, exclusively expressed phosphorylated, inactive PTEN, and active Akt isoforms, as did their downstream molecules. Among the Akt isoforms, enhanced expression of Akt1 was more prominent than that of Akt2 in both capsular and parenchymal carcinomas. CONCLUSIONS: Activation of the PI3K pathway through phosphorylation of PTEN promotes the high production of capsular carcinomas as well as the development of less frequent parenchymal carcinomas.


Subject(s)
PTEN Phosphohydrolase/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/drug effects , Sulfadimethoxine/toxicity , Thyroid Neoplasms/etiology , Animals , Cell Proliferation , Immunohistochemistry , Male , Neoplasm Invasiveness , PTEN Phosphohydrolase/analysis , Proto-Oncogene Proteins c-akt/analysis , Rats , Rats, Inbred F344 , Thyroid Neoplasms/chemistry , Thyroid Neoplasms/pathology
12.
Toxicol Appl Pharmacol ; 246(3): 128-40, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20423715

ABSTRACT

To clarify the involvement of signaling of transforming growth factor (TGF)-ß during the hepatocarcinogenesis, the immunohistochemical distribution of related molecules was analyzed in relation with liver cell lesions expressing glutathione S-transferase placental form (GST-P) during liver tumor promotion by fenbendazole, phenobarbital, piperonyl butoxide, or thioacetamide, using rats. Our study focused on early-stage promotion (6weeks after starting promotion) and late-stage promotion (57weeks after starting promotion). With regard to Smad-dependent signaling, cytoplasmic accumulation of phosphorylated Smad (phospho-Smad)-2/3 - identified as Smad3 by later immunoblot analysis - increased in the subpopulation of GST-P(+) foci, while Smad4, a nuclear transporter of Smad2/3, decreased during early-stage promotion. By late-stage promotion, GST-P(+) lesions lacking phospho-Smad2/3 had increased in accordance with lesion development from foci to carcinomas, while Smad4 largely disappeared in most proliferative lesions. With regard to Smad-independent mitogen-activated protein kinases, GST-P(+) foci that co-expressed phospho-p38 mitogen-activated protein kinase increased during early-stage promotion; however, p38-downstream phospho-activating transcriptional factor (ATF)-2, ATF3, and phospho-c-Myc, were inversely downregulated without relation to promotion. By late-stage promotion, proliferative lesions downregulated phospho-ATF2 and phospho-c-Myc along with lesion development, as with downregulation of phospho-p38 in all lesions. These results suggest that from the early stages, carcinogenic processes were facilitated by disruption of tumor suppressor functions of Smad-dependent signaling, while Smad-independent activation of p38 was an early-stage phenomenon. GST-P(-) foci induced by promotion with agonists of peroxisome proliferator-activated receptor-α did not change Smad expression, suggesting an aberration in the Smad-dependent signaling prerequisites for induction of GST-P(+) proliferative lesions.


Subject(s)
Carcinogenesis/pathology , Glutathione S-Transferase pi/metabolism , Liver Neoplasms, Experimental/pathology , Smad Proteins/metabolism , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogens/toxicity , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Genes, myc , Glutathione S-Transferase pi/genetics , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Male , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Inbred F344 , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad Proteins/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Chem Biol Interact ; 185(3): 189-201, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20302851

ABSTRACT

To investigate liver tumor promotion mechanisms of copper (Cu)- and iron (Fe)-overloading, immunolocalization of metal-related biomolecules and lipid peroxidation end products was examined in preneoplastic liver cell foci that expressed glutathione S-transferase placental form (GST-P) in early-stage tumor promotion over 6 weeks in a rat two-stage hepatocarcinogenesis model. Gene expression and concentrations of thiobarbituric acid-reactive substance (TBARS) in the liver were also analyzed. Cu-overloading alone exerted a weak promoting activity, which was enhanced by Fe-overloading. By Cu-overloading, GST-P(+) foci that co-expressed transferrin receptors or downregulated ceruloplasmin increased, suggesting preneoplastic lesion-specific enhancement of oxidative cellular stress. Cu-overloading also increased transcripts of antioxidant enzymes (Gstm3 and Gst Yc2 subunit), cell proliferation, and numbers of single liver cells expressing GST-P or heme oxygenase-1 (HO-1) in the liver, suggesting that oxidative stress induces single-cell toxicity, with the ensuing regeneration contributing to tumor promotion. Fe-overloading increased liver TBARS and HO-1-expressing Kupffer cells, the latter suggesting protection against inflammatory stimuli causing fluctuating proinflammatory cytokine mRNA levels. By co-overloading of Cu and Fe, Cu-overload-related single liver cell toxicity and regeneration increased, as did cytokine imbalances involving increased cyclooxygenase-2-producing Kupffer cells and accumulation of malondialdehyde within GST-P(+) foci. These results suggest an involvement of oxidative stress responses in Cu-induced tumor promotion and Fe-induced enhancement by increasing cytokine imbalances and GST-P(+) foci-specific lipid peroxidation.


Subject(s)
Copper/metabolism , Iron Overload/metabolism , Iron/metabolism , Liver Neoplasms, Experimental/metabolism , Oxidative Stress , Animals , Carcinogenicity Tests , Carcinogens/toxicity , Diethylnitrosamine/toxicity , Glutathione Transferase/metabolism , Immunohistochemistry , Liver/enzymology , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/pathology , Male , Rats , Rats, Inbred F344 , Thiobarbituric Acid Reactive Substances/analysis , Thiobarbituric Acid Reactive Substances/metabolism , Time Factors
14.
Reprod Toxicol ; 29(4): 407-14, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20347957

ABSTRACT

To detect molecular evidence reflecting a permanent disruption of neuronal development due to hypothyroidism, distribution of Reelin-producing cells that function in neuronal migration and positioning was analyzed in the hippocampal dentate hilus using rats. From gestation day 10, maternal rats were administered either 6-propyl-2-thiouracil (PTU) at 3 or 12ppm (0.57 or 1.97mg/kg body weight/day) or methimazole (MMI) at 200ppm (27.2mg/kg body weight/day) in the drinking water and male offspring were immunohistochemically examined at the end of exposure on weaning (postnatal day 20) and at the adult stage (11-week-old). Offspring with MMI and 12ppm PTU displayed evidence of growth retardation lasting into the adult stage. On the other hand, all exposure groups showed a sustained increase in Reelin-expressing cells in the dentate hilus until the adult stage in parallel with Calbindin-D-28K-expressing cells at weaning and with glutamic acid decarboxylase 67-positive cells in the adult stage, confirming an increase in gamma-aminobutyric acid (GABA)ergic interneurons. At the adult stage, NeuN-positive postmitotic mature neurons were also increased in the hilus in all exposure groups, however, the increased population of Reelin-producing cells at this stage was either weakly positive or negative for NeuN, indicative of immature neurons. At weaning, neuroblast-producing subgranular zone of the dentate gyrus showed increased apoptosis and decreased cell proliferation suggestive of impaired neurogenesis. The results suggest that sustained increases of immature GABAergic interneurons synthesizing Reelin in the hilus could be a signature of compensatory regulation for impaired neurogenesis and mismigration during the neuronal development as a hypothyroidism-related brain effect rather than that secondary to systemic growth retardation.


Subject(s)
Antithyroid Agents/toxicity , Cell Adhesion Molecules, Neuronal/metabolism , Dentate Gyrus/drug effects , Extracellular Matrix Proteins/metabolism , Interneurons/drug effects , Methimazole/toxicity , Nerve Tissue Proteins/metabolism , Propylthiouracil/toxicity , Serine Endopeptidases/metabolism , Animals , Calbindins , Cell Movement/drug effects , Cell Proliferation/drug effects , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Female , Glutamate Decarboxylase/metabolism , Interneurons/metabolism , Interneurons/pathology , Male , Maternal Exposure , Peptide Fragments , Pregnancy , Rats , Rats, Sprague-Dawley , Reelin Protein , S100 Calcium Binding Protein G/metabolism , gamma-Aminobutyric Acid/metabolism
15.
J Toxicol Sci ; 35(1): 69-78, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20118626

ABSTRACT

To determine the threshold dose of dicyclanil (DC) that induces hepatocellular tumor-promoting effects associated with reactive oxygen species (ROS) generation via their metabolic pathways, partial hepatectomized ICR male mice were fed diets containing 0, 187.5, 375 or 750 ppm DC after an intraperitoneal injection of N-diethylnitrosamine (DEN) to initiate hepatocarcinogenesis. Immunohistochemically, the proliferating cell nuclear antigen (PCNA)-positive cell ratio was significantly increased in the DEN + 750 ppm DC group compared with the DEN alone group. However, significant increases in the number of gamma-glutamyltranspeptidase (GGT)-positive cells and formation of microsomal ROS were not observed in the DEN + DC groups compared with the DEN alone group. Real-time polymerase chain reaction (RT-PCR) showed that the expression of Cyp1a1, Cyp1a2, and OGG1genes was significantly up-regulated in mice given diets containing 375 ppm DC or more, 187.5 ppm DC or more, and 750 ppm DC, respectively. These results suggest that the threshold dose of DC that induces ROS-mediated liver tumor promotion in mice is more than 750 ppm, although expression of the Cyp1a2 gene, which is related to ROS generation, was up-regulated in the liver of mice, even at a DC dose of 187.5 ppm.


Subject(s)
Carcinogens/toxicity , Carcinoma, Hepatocellular/chemically induced , Juvenile Hormones/toxicity , Liver Neoplasms/chemically induced , Animals , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/genetics , DNA Glycosylases/genetics , Diethylnitrosamine/toxicity , Dose-Response Relationship, Drug , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic/drug effects , Hepatectomy , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred ICR , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Reactive Oxygen Species , Up-Regulation/drug effects , gamma-Glutamyltransferase/metabolism
16.
Arch Toxicol ; 84(4): 319-31, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20091025

ABSTRACT

To elucidate the role of metal-related molecules in hepatocarcinogenesis, we examined immunolocalization of transferrin receptor (Tfrc), ceruloplasmin (Cp) and metallothionein (MT)-1/2 in relation to liver cell foci positive for glutathione-S-transferase placental form (GST-P) in the early stage of tumor promotion by fenbendazole (FB), phenobarbital, piperonyl butoxide or thioacetamide in a rat two-stage hepatocarcinogenesis model. To estimate the involvement of oxidative stress responses to the promotion, immunolocalization of 4-hydroxy-2-nonenal, malondialdehyde and acrolein was similarly examined. Our findings showed that MT-1/2 immunoreactivity was not associated with the cellular distribution of GST-P and proliferating cell nuclear antigen, suggesting no role of MT-1/2 in hepatocarcinogenesis. We also found enhanced expression of Tfrc after treatment with strong tumor-promoting chemicals. With regard to Cp, the population showing down-regulation was increased in the GST-P-positive foci in relation to tumor promotion. Up-regulation of Tfrc and down-regulation of Cp was maintained in GST-P-positive neoplastic lesions induced after long-term promotion with FB, suggesting the expression changes occurring downstream of the signaling pathway involved in the formation of GST-P-positive lesions. Furthermore, enhanced accumulation of lipid peroxidation end products was observed in the GST-P-positive foci by promotion. Post-initiation treatment with peroxisome proliferator-activated receptor alpha agonists did not enhance any such distribution changes in GST-P-negative foci. The results thus suggest that facilitation of lipid peroxidation is involved in the induction of GST-P-positive lesions by tumor promotion from an early stage, and up-regulation of Tfrc and down-regulation of Cp may be a signature of enhanced oxidative cellular stress in these lesions.


Subject(s)
Ceruloplasmin/metabolism , Glutathione S-Transferase pi/metabolism , Lipid Peroxidation , Liver Neoplasms, Experimental/metabolism , Liver/metabolism , Receptors, Transferrin/metabolism , Animals , Carcinogenicity Tests , Carcinogens , Copper/metabolism , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Neoplastic , Iron/metabolism , Liver Neoplasms, Experimental/etiology , Male , Metallothionein/metabolism , Oxidative Stress , PPAR alpha/metabolism , Rats , Rats, Inbred F344 , Up-Regulation
17.
Arch Toxicol ; 84(2): 155-64, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20101389

ABSTRACT

Piperonyl butoxide (PBO) is a pesticide synergist used with pyrethroids as a domestic insecticide, and it acts as a non-genotoxic hepatocarcinogen in rats and mice. To clarify whether oxidative stress is involved in the liver tumor-promoting effect of PBO in mice, male mice were subjected to two-thirds partial hepatectomy, followed by N-diethylnitrosamine (DEN) treatment, and given a diet containing 0.6% PBO for 25 weeks. The incidences of cytokeratin (CK) 8/18-positive foci, adenomas, and carcinomas significantly increased in the DEN + PBO group compared with the DEN-alone group. The PCNA-positive ratio significantly increased in non-tumor hepatocytes, CK8/18-positive foci and adenomas in the DEN + PBO group compared with the DEN-alone group. PBO increased reactive oxygen species (ROS) production in microsomes but did not change oxidative DNA damage as assessed by 8-hydroxydeoxyguanosine (8-OHdG). In real-time RT-PCR, PBO upregulated the expression of genes related to metabolism, such as Cytochrome P450 1a1, 2a5, and 2b10, and metabolic stress, such as Por and Nqo1, but downregulated Egfr and Ogg1. PBO also increased early response genes downstream of mitogen-activated protein kinase (MAPK), such as c-Myc that is induced by excessive ROS production, and G1/S transition-related genes, such as E2f1 and Ccnd1. Thus, PBO can generate ROS via the metabolic pathway without any induction of oxidative DNA damage, activate cell growth, increase c-Myc- and E2F1-related pathways, and act as a liver tumor promoter of DEN-induced hepatocarcinogenesis in mice.


Subject(s)
Cell Proliferation/drug effects , Liver Neoplasms, Experimental/chemically induced , Liver/drug effects , Pesticide Synergists/pharmacology , Piperonyl Butoxide/pharmacology , Animals , Body Weight/drug effects , Immunohistochemistry , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/metabolism , Male , Mice , Mice, Inbred ICR , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Organ Size/drug effects , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism
18.
Arch Toxicol ; 84(2): 143-53, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20033131

ABSTRACT

To clarify whether enzymatically modified isoquercitrin (EMIQ) or melatonin (MLT) supplementation reduces oxidative stress-mediated hepatocellular tumor-promoting effect of oxfendazole (OX), a benzimidazole anthelmintic, male rats were administered a single intraperitoneal injection of N-diethylnitrosamine (DEN) and were fed a diet containing OX (500 ppm) for 10 weeks with or without EMIQ (2,000 ppm) or MLT (100 ppm) in the drinking water after DEN initiation. One week after the commencement of the administration of OX, rats were subjected to two-thirds of partial hepatectomy. The number of GST-P-positive foci promoted by OX was significantly inhibited by the combined antioxidant EMIQ or MLT administration, and the area of GST-P-positive foci was inhibited by the administration of MLT. Real-time RT-PCR analysis revealed decreases in mRNA expression levels of cytochrome P450, family 2, subfamily b, polypeptide 2 (Cyp2b2) and malic enzyme 1 (Me1) in the DEN-OX-EMIQ and DEN-OX-MLT groups and decreases in mRNA expression levels of Cyp1a1 and aldo-keto reductase family 7, member A3 (Akr7a3) in the DEN-OX-MLT group compared to those in the DEN-OX group. In in vitro ROS production assay, inhibited production of NADPH-dependent ROS was observed by the treatment with EMIQ or MLT. These results suggest that coadministration of EMIQ or MLT suppresses the hepatocellular tumor-promoting activity of OX in rats through the decrease in ROS production by the activation of CYPs.


Subject(s)
Benzimidazoles/pharmacology , Carcinogens/pharmacology , Liver Neoplasms, Experimental/metabolism , Melatonin/metabolism , Quercetin/analogs & derivatives , Animals , Antioxidants/metabolism , Dietary Supplements , Hepatectomy , Liver Neoplasms, Experimental/pathology , Male , Oxidative Stress/drug effects , Quercetin/metabolism , Rats , Rats, Inbred F344
19.
J Vet Med Sci ; 72(3): 263-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20035116

ABSTRACT

In order to clarify whether cytokeratin (CK) 8/18 is a useful immunohistochemical marker for hepatocellular proliferative lesions in mice, partially hepatectomized male ICR mice were given 0.6% piperonyl butoxide (PBO) for 8 (Experiment I) or 25 weeks (Experiment II) after N-diethylnitrosamine (DEN) initiation treatment, and the livers were subjected to histological examinations on hematoxylin and eosin (HE) stained sections, CK8/18 immunohistochemistry and gamma-glutamyl transpeptidase (GGT) histochemistry. In Experiment I, the multiplicity of hepatocellular foci in paraffin-embedded sections which were observed in HE-stained sections and positive for CK8/18 was 10.17 and 18.50, respectively, while that of hepatocellular foci in frozen sections which were observed in HE-stained sections and positive/negative for GGT was 6.17 and 8.17, respectively. In Experiment II, the total multiplicity of hepatocellular foci in paraffin-embedded sections which were observed in HE-stained sections and positive/negative for CK8/18 was 4.47 and 23.17, respectively, while that of hepatocellular foci in frozen sections which were observed in HE-stained sections and positive/negative for GGT was 2.50 and 3.50, respectively. Most of the hepatocellular adenomas and carcinomas observed in HE-stained sections were positive for CK8/18, but some of the adenomas were negative for CK8/18. These findings indicate that more hepatocellular proliferative lesions can be detected in CK8/18 immunohistochemistry in addition to those observed in HE-stained sections, and suggest that CK8/18 may become a useful immunohistochemical marker for detecting hepatocellular proliferative lesions in mice.


Subject(s)
Carcinoma, Hepatocellular/pathology , Keratin-18/analysis , Liver Neoplasms, Experimental/pathology , Adenoma/chemically induced , Adenoma/pathology , Animals , Biomarkers/analysis , Carcinoma/chemically induced , Carcinoma/pathology , Carcinoma, Hepatocellular/chemically induced , Diethylnitrosamine , Hepatectomy , Immunohistochemistry , Keratin-8/analysis , Liver Neoplasms, Experimental/chemically induced , Male , Mice , Mice, Inbred ICR , Neoplasm Staging , Piperonyl Butoxide , Rats
20.
Exp Toxicol Pathol ; 62(3): 269-80, 2010 May.
Article in English | MEDLINE | ID: mdl-19505811

ABSTRACT

To investigate the cell cycle kinetics during the tumor promotion process induced by hypothyroidism in a rat model of thyroid follicular cell carcinogenesis, immunohistochemical analysis of cell cycle molecules and related signaling molecules was performed in conjunction with analysis of cell proliferation activity in an initiation-promotion model. Male F344 rats were injected with N-bis(2-hydroxypropyl)nitrosamine, and one week later treated with 6-propyl-2-thiouracil (PTU) at 12ppm in the drinking water for 4, 10 or 15 weeks. At each time point, proliferative lesions increased the expression of cyclin A, cyclin D, cyclin E and cyclin-dependent kinase (Cdk)-2, in association with the development of lesion stages from the early focal hyperplasia to the late carcinoma, while a subpopulation of proliferative lesions showed decreased numbers of both cell division cycle-2- and Ki-67-positive cells at week 15 compared with that at week 10, suggesting a reduced promoting effect of serum thyroid-stimulating hormone in the sensitive cellular population after long-term exposure to PTU. On the other hand, increased immunolocalization of phosphorylated and inactive glycogen synthase kinase (GSK)-3beta was observed in a subpopulation of proliferative lesions, in parallel with the cyclins and Cdk2. Nuclear immunoreactivity of phosphorylated and inactive retinoblastoma (Rb) protein was also increased in association with lesion development, with carcinomas showing increased cytoplasmic localization. The results suggest that proliferative lesions activate the cell cycle machinery following tumor promotion via a regulatory mechanism involving inactivation of GSK3beta and Rb protein, the latter signaling mechanism involving its aberrant nucleocytoplasmic transport for the acquisition of a malignant phenotype.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Glycogen Synthase Kinase 3/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction/physiology , Thyroid Neoplasms/metabolism , Adenocarcinoma, Follicular/chemically induced , Adenocarcinoma, Follicular/metabolism , Adenoma/chemically induced , Adenoma/metabolism , Animals , Carcinogens/toxicity , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/drug effects , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/chemically induced , Cytoplasm/metabolism , Disease Models, Animal , Glycogen Synthase Kinase 3 beta , Immunohistochemistry , Male , Nitrosamines/toxicity , Protein Transport/physiology , Rats , Rats, Inbred F344 , Thiouracil/toxicity , Thyroid Neoplasms/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...