Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 935: 175321, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36228744

ABSTRACT

Curcumin (aglycone curcumin) has antitumor properties in a variety of malignancies via the alteration of multiple cancer-related biological pathways; however, its clinical application has been hampered due to its poor bioavailability. To overcome this limitation, we have developed a synthesized curcumin ß-D-glucuronide sodium salt (TBP1901), a prodrug form of aglycone curcumin. In this study, we aimed to clarify the pharmacologic characteristics of TBP1901. In ß-glucuronidase (GUSB)-proficient mice, both curcumin ß-D-glucuronide and its active metabolite, aglycone curcumin, were detected in the blood after TBP1901 injection, whereas only curcumin ß-D-glucuronide was detected in GUSB-impaired mice, suggesting that GUSB plays a pivotal role in the conversion of TBP1901 into aglycone curcumin in vivo. TBP1901 itself had minimal antitumor effects in vitro, whereas it demonstrated significant antitumor effects in vivo. Genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 screen disclosed the genes associated with NF-κB signaling pathway and mitochondria were among the highest hit. In vitro, aglycone curcumin inhibited NF-kappa B signaling pathways whereas it caused production of reactive oxygen species (ROS). ROS scavenger, N-acetyl-L-cysteine, partially reversed antitumor effects of aglycone curcumin. In summary, TBP1901 can exert antitumor effects as a prodrug of aglycone curcumin through GUSB-dependent activation.


Subject(s)
Curcumin , Prodrugs , Animals , Mice , Cell Line, Tumor , CRISPR-Cas Systems/genetics , Curcumin/pharmacology , Glucuronidase/metabolism , Glucuronides/metabolism , Glucuronides/pharmacology , Glucuronides/therapeutic use , NF-kappa B/metabolism , Prodrugs/pharmacology , Prodrugs/therapeutic use , Reactive Oxygen Species/metabolism
2.
Cancer Sci ; 111(5): 1785-1793, 2020 May.
Article in English | MEDLINE | ID: mdl-32163218

ABSTRACT

The NF-kappa B (NF-κB) pathway plays a pivotal role in tumor progression and chemoresistance, and its inhibition has been shown to suppress tumor growth in a variety of preclinical models. Recently, we succeeded in synthesizing a water-soluble injectable type of curcumin ß-D-glucuronide (CMG), which is converted into a free-form of curcumin by ß-glucuronidase in vivo. Herein, we aimed to clarify the efficacy, safety and pharmacokinetics of CMG in a xenograft mouse model. First, we confirmed that the presence of KRAS/TP53 mutations significantly increased the IC50 of oxaliplatin (L-OHP) and NF-κB activity in HCT116 cells in vitro. Then, we tested the efficacy of CMG in an HCT116 colon cancer xenograft mice model. CMG demonstrated superior anticancer effects compared to L-OHP in an L-OHP-resistant xenograft model. With regard to safety, significant bodyweight loss, severe myelosuppression and AST/ALT elevation were observed in L-OHP-treated mice, whereas none of these toxicity was noted in CMG-treated mice. The combination of CMG and L-OHP exhibited additive effects in these xenograft models without increasing toxicity. Pharmacokinetic analysis revealed that high levels of free-form curcumin were maintained in the tumor tissue after 48 hours following CMG administration, but it was not detected in other major organs, such as the heart, liver and spleen. Immunohistochemistry revealed reduced NF-κB activity in the tumor tissue extracted from CMG-treated mice compared with that from control mice. These results indicated that CMG could be a promising anticancer prodrug for treating colon cancer with minimal toxicity.


Subject(s)
Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Curcumin/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Glucuronides/therapeutic use , Oxaliplatin/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Curcumin/chemistry , Curcumin/pharmacokinetics , Curcumin/pharmacology , Curcumin/therapeutic use , Female , Glucuronidase/metabolism , Glucuronides/chemistry , Glucuronides/pharmacokinetics , Glucuronides/pharmacology , HCT116 Cells , Humans , Mice , Mice, Nude , Mutation , NF-kappa B/metabolism , Oxaliplatin/therapeutic use , Prodrugs , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
3.
Sci Rep ; 9(1): 13405, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31527634

ABSTRACT

Various new drugs have been developed for treating recurrent hormone receptor-positive (HR+)/human epidermal receptor 2-negative (HER2-) breast cancer. However, directly identifying effective drugs remains difficult. In this study, we elucidated the clinical relevance of cultured cells derived from patients with recurrent HR+/HER2- metastatic breast cancer. The recently established conditionally reprogrammed (CR) cell system enables us to examine heterogeneity, drug sensitivity and cell function using patient-derived tumour samples. The results of microarray analysis, DNA target sequencing and xenograft experiments indicated that the mutation status and pathological features were preserved in CR cells, whereas RNA expression was different from that in the primary tumour cells, especially with respect to cell adhesion-associated pathways. The results of drug sensitivity assays involving the use of primary breast cancer CR cells were consistent with gene expression profiling test data. We performed drug-screening assays using liver metastases, which were sensitive to 66 drugs. Importantly, the result reflected the actual clinical course of this patient. These results supported the use of CR cells obtained from the metastatic lesions of patients with HR+/HER2- breast cancer for predicting the clinical drug efficacy.


Subject(s)
Breast Neoplasms/drug therapy , Cellular Reprogramming , Drug Resistance, Neoplasm/drug effects , Neoplasm Recurrence, Local/drug therapy , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis , Biological Assay , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Early Detection of Cancer/methods , Female , Gene Expression Profiling , High-Throughput Screening Assays/methods , Humans , Mice , Mice, Nude , Mutation , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Tamoxifen/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Heliyon ; 5(12): e03051, 2019 Dec.
Article in English | MEDLINE | ID: mdl-32083202

ABSTRACT

An earthquake struck the eastern part of Japan on March 11, 2011. The Fukushima Daiichi nuclear power plant was severely damaged by the earthquake and subsequent tsunami, leading to the emission of large amounts of radioactive pollutants, including 134Cs and 137Cs, into the environment. From August 23 to September 1 in 2011, and from August 27 to September 4 in 2013, we collected samples of animals, plants, fungi and lichens from Svalbard, Norway and measured the radioactivity of 134Cs and 137Cs contained in the samples. Though no radioactivity of 134Cs, which has a half-life of approximately 2 years, was observed, radioactivity of 137Cs, which has a half-life of approximately 30 years, was observed in some samples of lichens and fungi. We failed to detect the radioactivity of 134Cs in any of the samples we collected, therefore, it was impossible to say clearly that the radioactivity is derived from Fukushima or not. Nevertheless, the radioactivity data documented in this report are a useful reference for the future surveys of radioactivity within the Arctic.

5.
Anticancer Res ; 38(3): 1311-1316, 2018 03.
Article in English | MEDLINE | ID: mdl-29491054

ABSTRACT

BACKGROUND: CD147 (basigin/emmprin) is expressed on the surface of carcinoma cells. MATERIALS AND METHODS: For studying the efficacy of CD147-targeting medicine on CD147-expressing cells, we studied the effect of anti-CD147-labeled polymeric micelles (CD147ab micelles) that encapsulated a conjugate of doxorubicin with glutathione (GSH-DXR), with specific accumulation and cytotoxicity against CD147-expressing A431 human epidermoid carcinoma cells, Ishikawa human endometrial adenocarcinoma cells, and PC3 human prostate carcinoma cells. RESULTS: By treatment of each cell type with CD147ab micelles for 1 h, a specific accumulation of CD147ab micelles in CD147-expressing cells was observed. In addition, the cytotoxicity of GSH-DXR-encapsulated micelles against each cell type was measured by treatment of the micelles for 1 h. The cytotoxic effect of CD147ab micelles carrying GSH-DXR was 3- to 10-fold higher for these cells than that of micelles without GSH-DXR. CONCLUSION: These results suggest that GSH-DXR-encapsulated CD147ab micelles could serve as an effective drug delivery system to CD147-expressing carcinoma cells.


Subject(s)
Antibodies/chemistry , Basigin/metabolism , Doxorubicin/analogs & derivatives , Micelles , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Antibodies/immunology , Antibodies/pharmacology , Basigin/antagonists & inhibitors , Basigin/immunology , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/chemistry , Doxorubicin/pharmacology , Glutathione/analogs & derivatives , Glutathione/chemistry , Humans , Mice
6.
Int J Oncol ; 50(6): 2180-2190, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28498467

ABSTRACT

It has been proposed that α-fetoprotein (AFP) is a new member of the intracellular signaling molecule family of the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway via interaction with the phosphatase and tensin homolog (PTEN). In this study, the effects of anti-human AFP antibody on the functions of PTEN were examined using an AFP-producing human hepatoma cell line. The antibody caused significant inhibition of cell growth, compared to a normal IgG control, with the accumulation of intracellular immune complexes followed by significant reduction of cytosolic functional AFP. Decrease in the amount of AKT phosphorylated on serine (S) 473 indicated that PI3K/AKT signaling was suppressed in the cells. S380-phosphorylated PTEN increased markedly by the second day after antibody treatment, with slight but significant increase in the PTEN protein level. Since phosphorylation at S380 is critical for PTEN stability, the increase in S380-phosphorylated PTEN indicated maintenance of the number of PTEN molecules and the related potential to control PI3K/AKT signaling. p53 protein (P53) significantly, but slightly increased during antibody treatment, because PTEN expression increased the stability and function of P53 via both molecular interactions. P53 phosphorylated at S20 or at S392 dramatically increased, suggesting an increase in the stability, accumulation and activation of P53. Glucose transporter 1 (GLUT1) increased immediately after antibody treatment, pointing to a deficiency of glucose in the cells. Immunofluorescence cytology revealed that antibody-treatment re-distributed GLUT1 molecules throughout the cytoplasm with a reduction of their patchy localization on the cell surface. This suggested that translocation of GLUT1 depends on the PI3K/AKT pathway, in particular on PTEN expression. Antibody therapy targeted at AFP-producing tumor cells showed an inhibitory effect on the PI3K/AKT pathway via the liberation, restoration and functional stabilization of PTEN. PTEN simultaneously induced both P53 activation and intracellular translocation of GLUT1, since these are closely associated with PTEN.


Subject(s)
Carcinoma, Hepatocellular/therapy , Glucose Transporter Type 1/genetics , Liver Neoplasms/therapy , PTEN Phosphohydrolase/genetics , Tumor Suppressor Protein p53/genetics , alpha-Fetoproteins/genetics , Antibodies/administration & dosage , Antibodies/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glucose Transporter Type 1/immunology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/drug effects , Tumor Suppressor Protein p53/immunology , alpha-Fetoproteins/antagonists & inhibitors , alpha-Fetoproteins/immunology
7.
Int J Oncol ; 46(5): 2251-60, 2015 May.
Article in English | MEDLINE | ID: mdl-25738863

ABSTRACT

Downregulation of E-cadherin (gene: CDH1) plays an important role in epithelial-mesenchymal transition (EMT), which is critical for normal development and disease states. As a result of long-term treatment of endometrial carcinoma Ishikawa cells with epoxomicin (EXM), the cells exhibited the phenotype for EXM-resistance (Ish/EXM cells). Moreover, CDH1 mRNA and its protein were suppressed and EMT was induced in Ish/EXM cells. Ish/EXM cells exhibited drug-resistance to other proteasome inhibitors, MG-132, PSI and PS-341 (Bortezomib). The proteasome inhibitor-resistant cells acquired invasiveness as a result of the chemotherapy. In Ish/EXM cells, E-cadherin was suppressed by upregulation of its transcriptional repressor ZEB1. Furthermore, expression of the miR-200 family (miR-200a, miR-200b, miR-200c and miR-141) found in Ishikawa cells was suppressed in Ish/EXM cells. Overexpression of the miR-200 family in Ish/EXM cells caused by transfection with the pre-miR-200 family induced downregulation of ZEB1 and enhanced expression of E-cadherin. Conversely, suppression of miR-200 expression in the Ishikawa cells by transfection with anti-miR-200 elevated the expression of ZEB1 and suppressed the expression of E-cadherin. These results suggest that acquirement of EXM-resistance in Ish/EXM cells induces up regulation of ZEB1 via suppression of the miR-200 family following suppression of E-cadherin. Since suppression of ZEB1 in Ish/EXM cells by treatment with its siRNA did not restore the miR-200 family expression, miR-200 family was placed upstream of ZEB1 to regulate the expression.


Subject(s)
Cadherins/biosynthesis , Endometrial Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/genetics , MicroRNAs/genetics , Transcription Factors/genetics , Blotting, Western , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Humans , Proteasome Inhibitors/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Zinc Finger E-box-Binding Homeobox 1
8.
Int J Oncol ; 40(6): 2122-30, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22447231

ABSTRACT

Ovarian clear cell carcinoma (OCCC) has several significant characteristics based on molecular features that are distinct from those of ovarian high-grade serous carcinoma. Cellular glycogen accumulation is the most conspicuous feature of OCCC and in the present study its metabolic mechanism was investigated. The amount of glycogen in cells cultured under hypoxia increased significantly and approximately doubled after 48 h (P<0.01) compared to that under normoxic conditions. Periodic acid-Schiff positive staining also demonstrated intracellular glycogen storage. Western blot analysis revealed that HIF1α, which was overexpressed and stabilized under hypoxic conditions, led to an increase in the levels of cellular glycogen synthase 1, muscle type (GYS1), and conversely to a decrease in inactive phosphorylated GYS1 at serine (Ser) 641. Additional increases were observed in both protein phosphatase 1, which dephosphorylates and thereby induces GYS1 enzyme activity, and glycogen synthase kinase 3 beta (GSK3ß) phosphorylated at Ser9, which is inactive on phosphorylation of GYS1 and subsequently induces its enzyme activity. By contrast, the level of PYGM-b decreased. These results indicated that the glycogen accumulation under a hypoxic environment resulted in the promotion of glycogen synthesis, but did not lead to inhibition of glycogen degradation and/or consumption. Under hypoxic conditions, HAC2 cells showed activation of the PI3K/AKT pathway caused by a mutation in exon 20 of PIK3CA, encoding the catalytic subunit p110α of PI3K. The resulting activation of AKT (phosphoSer473) also plays a role as a central enhancer in glycogen synthesis through suppression of GSK3ß via phosphorylation at Ser9. Hypoxia decreased the cytocidal activity of cisplatin and doxorubicin to various degrees. In conclusion, the hypoxic conditions together with HIF1 expression and stabilization increased the intracellular glycogen contents and resistance to the anticancer drugs.


Subject(s)
Carcinoma/metabolism , Glycogen/biosynthesis , Ovarian Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Base Sequence , Cell Hypoxia/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Class I Phosphatidylinositol 3-Kinases , DNA Mutational Analysis , Doxorubicin/pharmacology , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/genetics
9.
Int J Oncol ; 39(5): 1327-36, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21785823

ABSTRACT

In vitro tumor growth in a three-dimensional (3D) architecture has been demonstrated to play an important role in biology not only for developmental organogenesis and carcinogenesis, but also for analyses on reconstitution and maintenance in a variety of biological environments surrounding the cells. In addition to providing architectural similarity to living organisms, 3D culture with a radial flow bioreactor (RFB) can also closely mimic the living hypoxic microenvironment under which specific organogenesis or carcinogenesis occurs. The findings of the present study under the RFB culture conditions show that cancer cells underwent a shift from aerobic to hypoxic energy metabolism, in addition to protein expression to maintain the 3D structure. In RFB-cultured cells, protein stability of hypoxia-inducible factor 1 (HIF1) α, a subunit of HIF1, was increased without upregulation of its mRNA. Under these conditions, PHD2, HIF-prolyl-4-hydroxy-lase 2 and a HIF1 downstream enzyme, were stabilized without affecting the mRNA levels via downregulation of FK506-binding protein 8. PHD2 accumulation, which occurred concomitant with HIF1 stabilization, may have compensated for the lack of oxygen under hypoxic conditions to regulate the HIF levels. 3D-culture-induced overexpression of carbonic anhydrase (another representative HIF downstream enzyme) was found to occur independently of cell density in RFB--cultured cells, suggesting that the RFB provided an adequately hypoxic microenvironment for the cultured cells. From these results, it was hypothesized that the key factors are regulatory molecules, which stabilize and degrade HIF molecules, thereby activating the HIF1 pathway under a hypoxic milieu.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Microenvironment/drug effects , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Carcinoma, Squamous Cell/genetics , Cell Culture Techniques , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Line, Tumor , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Nude , Spheroids, Cellular , Squamous Cell Carcinoma of Head and Neck , Transcriptional Activation/genetics , Tumor Cells, Cultured , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
10.
Int J Oncol ; 36(1): 77-83, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19956835

ABSTRACT

It was confirmed that CD147 (Emmprin) was expressed on the cell surface of carcinoma cells. For the purpose of studying the efficacy of a CD147-targeting agent on CD147-expressing carcinoma cells, we investigated the effect of a conjugate of glutathione-doxorubicin (GSH-DXR) encapsulated in an anti-CD147 antibody-labeled liposome (aCD147ab-liposome) in terms of specific accumulation and cytotoxicity in CD147-expressing human carcinoma cells. Expression of CD147 was not observed in many normal human tissues. However, slight expression of CD147 in kidney, prostate and breast tissues was observed. By contrast, high-level expression of CD147 in all carcinoma cells such as A431, PC3 and Ishikawa cell lines was confirmed by fluorescent microscopy and Western blot analysis. Specific accumulation of the aCD147ab-liposome in the above-described CD147-expressing cells was observed. GSH-DXR encapsulated in an aCD147ab-liposome expressed specific cytotoxicity against these carcinoma cells. These results suggested that target chemotherapy of GSH-DXR encapsulated in an aCD147ab-liposome on CD147-expressing carcinoma cells was effective.


Subject(s)
Antibodies, Monoclonal/chemistry , Basigin/chemistry , Carcinoma/metabolism , Doxorubicin/analogs & derivatives , Gene Expression Regulation, Neoplastic , Glutathione Transferase/chemistry , Glutathione/analogs & derivatives , Liposomes/chemistry , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Doxorubicin/chemistry , Doxorubicin/pharmacology , Glutathione/chemistry , Glutathione/pharmacology , Humans , Mice , Time Factors , Tissue Distribution
11.
Int J Oncol ; 34(5): 1433-48, 2009 May.
Article in English | MEDLINE | ID: mdl-19360357

ABSTRACT

To confirm the usefulness of the radial flow type bioreactor (RFB) for a three-dimensional (3D) culture system, which provides a tissue architecture and molecular function mimicking the in vivo environment, molecular expression in the A431 human squamous carcinoma cell line during culture were analyzed under the physically different environments of 3D culture in the RFB, 2D culture in a monolayer as well as in nude mice. Time-dependent accumulation of autocrine transforming growth factor (TGF) beta1 was found in spent culture media obtained only from 3D cultured A431 cancer cells, which grew well with a stratified-sheet morphology. Cells in the RFB overexpressed matrix metalloproteinase 7 (MMP7) and showed an increased release of soluble 80-kDa fragments of E-cadherin into the media time-dependently, resulting in the reduction of E-cadherin protein at the cell surface without down-regulation of the mRNA. beta-Catenin and its nuclear partner, LEF1, were up-regulated and Wnt protein secretion was also accelerated. Additional up-regulation of the transcriptional factors, HMGA2 and down-stream Slug, was noted. TGFbeta1-dependent, MMP7-mediated up-regulation of beta-catenin/LEF1 signaling and TGFbeta1-activated HMGA2 pathways consequently converged with Slug overexpression, due to disassembly and further repression of E-cadherin expression, which was reproducible in the epithelial mesenchymal transition process without any manipulation. Other transcriptional factors, Notch/HEY1 and NF-kappaB, were also up-regulated in 3D-cultured cells. These signals recruited molecules related to extracellular matrix-cell remodeling and angiogenesis. Expression of several representative molecules in the 3D cultured cells was parallel with that in xenotransplanted A431 tumor tissues in nude mice. 3D culture of tumor cells in the RFB is a useful tool for cancer experimental biology and evaluation of cancer therapeutic-like systems in nude mice.


Subject(s)
Bioreactors , Cell Transdifferentiation/genetics , Matrix Metalloproteinase 7/physiology , Neoplasms/pathology , Smad Proteins/physiology , Transforming Growth Factor beta1/physiology , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Culture Techniques/methods , Cell Transdifferentiation/physiology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/physiology , Gene Expression Regulation, Neoplastic , Humans , Male , Matrix Metalloproteinase 7/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Xenograft Model Antitumor Assays
12.
Int J Oncol ; 33(2): 389-95, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18636161

ABSTRACT

We investigated the induction of apoptosis via deamidation of Bcl-xL and translocation of Bax to the mitochondria by treatment with GSH-DXR. GSH-DXR treatment of HepG2 cells, which did not express GST P1-1, exhibited deamidation of Bcl-xL, and the degree of deamidation was related to the activation of caspase-3. Overexpression of GST P1-1 in HepG2 cells decreased both the Bcl-xL deamidation and caspase-3 activation induced by treatment with GSH-DXR. Bcl-xL deamidation and caspase-3 activation were also suppressed by co-treatment with SP600125, a specific inhibitor of JNK activity. Overexpression of wild-type Bcl-xL in HepG2 decreased GSH-DXR-induced apoptosis although deamidation was observed. However, expression of the deamidated mutant of Bcl-xL, in which aspartic acid was substituted for both arginine 52 and 66 (N52,66D-Bcl-xL), exhibited high sensitivity for the induction of apoptosis. Expression of the Bcl-xL mutant, in which alanine was substituted for both arginine 52 and 66 (N52,66A-Bcl-xL), suppressed deamidation and showed resistance to the induction of apoptosis by treatment with GSH-DXR. On the other hand, endogenous Bax and overexpressed Flag-Bax were localized in the cytosolic fraction of HepG2 cells. Treatment of the cells with GSH-DXR caused translocation of Flag-Bax to the mitochondrial fraction following the induction of apoptosis. The induced apoptosis was enhanced by the expression of Flag-Bax. Moreover, Flag-Bax was partly located in the mitochondrial fraction in N52,66D-Bcl-xL-expressed cells without the induction of apoptosis. Therefore, the induction of apoptosis by treatment of HepG2 with GSH-DXR was enhanced, thereby facilitating the release of cytochrome c by both deamidated inactivation of Bcl-xL and functional translocation of Bax to the mitochondria via JNK activation. Deamidation of Bcl-xL might be induced in order to translocate Bax to the mitochondria.


Subject(s)
Apoptosis/physiology , Doxorubicin/analogs & derivatives , Glutathione/analogs & derivatives , MAP Kinase Kinase 4/metabolism , Mitochondria/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Apoptosis/drug effects , Blotting, Western , Caspase 3/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Glutathione/pharmacology , Humans , Protein Transport/drug effects , Protein Transport/physiology , bcl-X Protein/drug effects
13.
World J Gastroenterol ; 13(40): 5306-11, 2007 Oct 28.
Article in English | MEDLINE | ID: mdl-17879398

ABSTRACT

AIM: To investigate the expression of survivin during the early stages of hepatocellular carcinoma (HCC). METHODS: Immunohistochemical expression of survivin in liver tumor and non-tumor tissue specimens taken from 17 patients was compared. In addition, to determine the survivin expression in response to anti-cancer drugs in early stage HCC, the survivin expression was determined after the treatment of the HCC cells with anti-cancer drugs under hypoxic culture conditions. RESULTS: Survivin proteins were expressed in 64.7% of cells in early HCC specimens. A correlation between the survivin expression rate in the peritumoral hepatocytes and the rate of expression in the HCC specimens (low-rate group vs high-rate group) was observed. The survivin protein concentration in HCC cells was increased by the combination of hypoxia and anti-cancer drugs. CONCLUSION: This study suggests that survivin could be used as a therapeutic target in early HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Cell Hypoxia , Epirubicin/pharmacology , Liver Neoplasms/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Biopsy , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Epirubicin/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Microtubule-Associated Proteins/genetics , Middle Aged , Neoplasm Proteins/genetics , Survivin
14.
Apoptosis ; 12(7): 1269-80, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17431793

ABSTRACT

Treatment of cells with a synthetic conjugate of DXR with GSH via glutaraldehyde (GSH-DXR) caused cytochrome c to be released from the mitochondria to the cytosol following potent activation of caspase-3 and -9 by typical DNA fragmentation. This apoptosis was regulated by the JNK-signaling pathway. In the present experiment, binding of GSH-DXR to GST P1-1 allosterically led to the disappearance of its enzyme activity and activated the kinase activity of JNK without dissociation of the JNK-GST P1-1 complex. The recombinant GST P1-1 molecule with a mutation in the active center region (W38H and C47S) lost its GST activity when bound to JNK to the same degree as the wild-type, with the mutated GST P1-1 molecule failing to inhibit the activity of JNK. It has been reported that JNK-signaling is regulated by GST P1-1 via interaction with the C-terminus. We confirmed that GST P1-1 deletion mutant (Delta194-209) and a site-directed mutant (R201A) in the C-terminal region failed to bind and inhibit JNK. These results indicated that not only binding of the C-terminal region of GST P1-1 to the JNK molecule, but also the active center region of GST P1-1 play important roles in the regulation of JNK enzyme activity. The findings suggested that allosteric inhibition of GST P1-1 activity by the binding of GSH-DXR following conformational change may activate JNK and induce apoptosis via the mitochondrial pathway in the cells.


Subject(s)
Apoptosis , Doxorubicin/analogs & derivatives , Glutathione S-Transferase pi/metabolism , Glutathione/analogs & derivatives , JNK Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Doxorubicin/metabolism , Doxorubicin/pharmacology , Enzyme Activation , Glutathione/metabolism , Glutathione/pharmacology , Glutathione S-Transferase pi/antagonists & inhibitors , Glutathione S-Transferase pi/chemistry , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mutation , Rats , Recombinant Fusion Proteins/metabolism , Transfection
15.
Oncol Rep ; 14(3): 601-7, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16077962

ABSTRACT

The relationship between the expression level of putative drug resistance factors and sensitivity to anticancer drugs in human normal renal proximal tubule epithelial cells (RPTEC) and 3 kinds of renal cell carcinoma (RCC) cells, VMRC-RCW (RCW), OS-RC-2 (OS2), TUHR14TKB (14TKB), was examined. RPTEC exhibited high expression of P-glycoprotein (Pgp), gamma-glutamyl cysteine synthetase (gammaGCS) and cis-diamminedichloroplatinum (II) (CDDP) resistance-related gene 9 (CRR9), low expression of vacuolar ATPase (V-ATPase) and no expression of multidrug resistance-associated protein 1 (MRP1). 14TKB exhibited high expression of gammaGCS and CRR9, low expression of Pgp and V-ATPase, and no expression of MRP1. OS2 showed high expression of CRR9, low expression of Pgp, gammaGCS and MRP1, and no expression of V-ATPase. RCW exhibited high expression of Pgp, MRP1 and CRR9 and low expression of gammaGCS and V-ATPase. The level of expression of the resistance factors varied among the cells. GST activity and GST-pi expression level of each cell were correlated, and there were high levels in OS2 and RPTEC. When the cytotoxicity of anticancer drugs against each cell was measured at 96 h, the sensitivity to CDDP and Doxorubicin (DXR) in RPTEC and RCW was lower than that in the other cells. Sensitivity to DXR was enhanced by treatment with the Pgp inhibitor, Verapamil, in proportion to the Pgp expression level, and the sensitivity to CDDP was increased by the gammaGCS inhibitor, Buthionine sulfoximine, in proportion to the gammaGCS expression level (corresponding to GSH content). Although a significant increase in sensitivity to CDDP was not observed by treatment of RCC with the V-ATPase inhibitor, Bafilomycin, the sensitivity to DXR in Bafilomycin-treated cells increased about 2-fold. However, no relation between drug sensitivity and V-ATPase expression was observed. The features (such as degree of resistance) varied among the RCC cell lines manifesting many resistance factors or to the contrary, lacking or having lowered resistance factors in comparison with normal cells. Therefore, it is necessary in clinical cancer chemotherapy to determine and measure the level of expression of each resistance factor in respective tumor tissue.


Subject(s)
Carcinoma, Renal Cell/genetics , Epithelial Cells/metabolism , Kidney Neoplasms/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Resistance/genetics , Epithelial Cells/cytology , Epithelial Cells/drug effects , Gene Expression/drug effects , Glutamate-Cysteine Ligase/genetics , Glutathione/metabolism , Glutathione Transferase/metabolism , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Tubules, Proximal/cytology , Membrane Proteins/genetics , Multidrug Resistance-Associated Proteins/genetics , Neoplasm Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vacuolar Proton-Translocating ATPases/genetics
16.
Curr Cancer Drug Targets ; 4(7): 577-90, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15578915

ABSTRACT

In cancer chemotherapy, it is necessary to design an agent that suppresses or inhibits the targets that influence cell growth and apoptosis. We focus on the apoptotic pathway via mitochondria in this article. In this pathway, c-Jun N-terminal kinase (JNK), one of the stress activated protein kinases, is predominantly activated by apoptotic stimuli. JNK activity is inhibited by the binding of glutathione S-transferase P1-1 (GST P1-1) through protein-protein interactions. It has been noted that GST P1-1 overexpression plays an important role in carcinogenesis and in part in the MDR phenotype. We show several useful modifications of an anticancer agent that suppress the enzyme activity and expression of GST P1-1. The release of cytochrome c from mitochondria to the cytosol during apoptosis is mediated by the mitochondrial permeability transition pore, which is a protein complex formed by the voltage-dependent anion channel, members of the pro- and anti- apoptotic Bax-Bcl-2 protein family, cyclophilin D, and adenine nucleotide (ADP/ATP) translocators. We propose some drugs, including a proteasome inhibitor that can triger the permeability transition.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Mitochondria/drug effects , Animals , Apoptosis/physiology , DNA Fragmentation/drug effects , DNA Fragmentation/physiology , Humans , Mitochondria/physiology
17.
Int J Oncol ; 24(2): 425-33, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14719120

ABSTRACT

A431 resistant variants to epoxomicin (EXM) were established, showing 4.0-6.7 times more resistance to EXM than parental A431P. Both variants demonstrated increased expression of the beta-subunit molecules of 26S proteasome with approximately 2.5 times increased activity. In variant cells, cyclin B and P34cdc2 were over-expressed, whereas P21WAF1 was expressed at a similar level to A431P. Because of the proteasome inhibitor acting as a G2/M blocker, results are to the advantage of resistant cells proliferating in the presence of an inhibitor under a severe environment. Variant cells showed increased expression of epidermal growth factor receptor (EGFR) and decreased expression of mRNA, but also slight accumulation of protein of c-Cbl, which is a negative regulator of EGFR possessing ubiquitin ligase activity to desensitize EGF signaling. UbcH7, acting intimately with c-Cbl, was decreased in level compared to A431P. These phenomena can be regarded as one of the causes of prevention of c-Cbl-mediated down-regulation of EGFR in variant cells, enabling them to live. The anti-apoptotic Bcl-2 mainly consisted of a phosphorylated form with resistance to proteasomal degradation, suggesting that Bcl-2 phosphorylation occurred independently of its apoptotic function. Variant cells showed resistance not only to EXM, but to the 5 proteasome inhibitors, while demonstrating collateral sensitivity to doxorubicin.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Multienzyme Complexes/antagonists & inhibitors , Oligopeptides/pharmacology , Apoptosis , Blotting, Western , CDC2 Protein Kinase/metabolism , Caspase 3 , Caspases/metabolism , Cell Division , Cell Line, Tumor , Cyclin B/metabolism , Cysteine Endopeptidases , Dose-Response Relationship, Drug , Down-Regulation , Electrophoresis, Polyacrylamide Gel , ErbB Receptors/metabolism , G2 Phase , Humans , Inhibitory Concentration 50 , Mitosis , Peptide Hydrolases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-cbl , RNA, Messenger/metabolism , Signal Transduction , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism
18.
Alcohol Clin Exp Res ; 26(8 Suppl): 32S-37S, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12198372

ABSTRACT

BACKGROUND: A carbohydrate-deficient transferrin (CDT) is the most useful marker of alcohol abuse; however, the mechanism of production and the pathophysiologic roles of CDT remain obscure. The effects of alcohol and its metabolites on growth and proliferation, transferrin synthesis, and phosphomannomutase enzyme activity in a human hepatoblastoma, HepG2, were examined. METHODS: HepG2 cells were treated with either ethanol at 80 mM or acetaldehyde at 400 microM. Transferrin secreted by the cells was prepared from conditioned culture medium by single-step immunoaffinity column chromatography using a goat-specific antibody against human transferrin. Phosphomannomutase and some related enzyme activities in the cell extracts were determined. Reverse transcription-polymerase chain reaction analysis of phosphomannomutase mRNA expression was also determined in HepG2 cultured with or without acetaldehyde (400 microM). RESULTS: HepG2 cells usually synthesized and secreted transferrin with three separated bands: main broad bands estimated to be 78 to 82 kDa, 75 kDa, and 72 kDa. The last two bands were compatible with part or the entire N-glycans-deficient transferrin (CDT) from alcoholic liver damage. Increased secretion of CDT from HepG2 correlated well with the extent of growth retardation to the level of confluent cell density. The activity of phosphomannomutase also decreased with prolongation of cellular doubling time. Furthermore, acetaldehyde treatment at 400 microM accelerated the inhibitory effect of cell growth compared with nontreated cells, and this condition facilitated CDT secretion from HepG2 cells. Determination of the enzyme activity and mRNA expression indicated that acetaldehyde showed competitive type inhibition of phosphomannomutase activity but not suppression of phosphomannomutase gene expression. CONCLUSIONS: By culturing HepG2 cells with acetaldehyde containing media, growth inhibition-dependent increase of CDT showed good correlation with reduced enzyme activity of phosphomannomutase. Acetaldehyde facilitated growth retardation, inhibition of phosphomannomutase activity, and increased secretion of CDT. The HepG2 cell line is useful as an in vitro model to investigate the pathophysiologic state of alcoholic liver damage and mechanisms of production as well as the physiologic role of CDT.


Subject(s)
Cell Division/drug effects , Ethanol/toxicity , Hepatocytes/drug effects , Liver Diseases, Alcoholic/enzymology , Phosphotransferases (Phosphomutases)/antagonists & inhibitors , Transferrin/analogs & derivatives , Transferrin/metabolism , Carcinoma, Hepatocellular , Cell Count , Cell Division/genetics , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/enzymology , Humans , Liver Diseases, Alcoholic/genetics , Liver Neoplasms , Phosphotransferases (Phosphomutases)/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...