Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Chem Biol Interact ; 369: 110257, 2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36375514

ABSTRACT

Compounds with 3,4-fused tricyclic indole (FTI) frameworks are attractive scaffolds for drug discovery. We synthesized FTI-6D, a compound with this framework, which was cytotoxic in several human cancer cell lines. FTI-6D induced apoptosis via activation of the p53 downstream mitochondria-related apoptotic pathway, characterized by an increased ratio of pro-apoptotic Bcl-2 family members to anti-apoptotic members. This change was followed by caspase-9 and caspase-3 cleavage and activation in two cancer cell lines, RKO and AGS. The anti-proliferating effect of FTI-6D was remarkably detected in eight cancer cells with wild-type TP53 (TP53_wt), including RKO and AGS, but not in seven cancer cells with mutated TP53 (TP53_mut). Additionally, p53 protein levels increased after FTI-6D treatment in TP53_wt cancer cells, and the cytotoxic effect of FTI-6D was decreased by TP53 knockdown. Accordingly, the expression of p53 downstream genes involved in apoptotic signaling pathways, such as BBC3 and TP53INP1, and those involved in cell growth inhibition, such as CDKN1A, was upregulated in TP53_wt cancer cells. These results suggest that the anti-proliferative and apoptosis-inducing activities of FTI-6D rely on p53 and the corresponding signaling processes. This study demonstrated that FTI-6D shows anti-cancer activity against TP53_wt cancer cells. FTI-6D may have potential as a prototype compound for a new drug to utilize a functional p53 pathway in TP53_wt cancer cells.


Subject(s)
Neoplasms , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Genes, p53 , Apoptosis , Cell Line, Tumor , HCT116 Cells , Neoplasms/genetics , Carrier Proteins/genetics , Heat-Shock Proteins/metabolism
2.
Biomed Rep ; 13(4): 22, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32765861

ABSTRACT

Lifeceramics (LC) is made of zeolite and oyster shell and is hypothesized to act as an anti-oxidative agent. In the present study, the effects of LC-treated water (LC water) on the concentration of serum uric acid (SUA) and the hemorheological parameters in male rats with hyperuricemia (HUA) was assessed. To prepare LC water, distilled water was mixed with LC particles. HUA was induced in rats by daily potassium oxonate (PO) injection (250 mg/kg). The PO-injected rats were separated into three different groups and were administered distilled water (PO rats), allopurinol [a xanthine oxidase (XOD) inhibitor] solution [PO + allopurinol (AP) rats] or LC water (PO+LC rats) by gavage. Control rats were intraperitoneally injected with sodium carboxymethyl cellulose solution and administered untreated distilled water by gavage. After injection and gavage for 5 weeks, the SUA concentration, hemorheology index and antioxidant index were measured. The SUA concentration and blood deformation index of the PO rats were significantly higher and lower, respectively, compared with the control rats. However, in the PO+LC rats, the SUA concentration and blood deformation index decreased and increased, respectively, to a level similar to that of the control as well as that in the PO+AP rats. Furthermore, the PO-induced increase in XOD activity was suppressed by combined treatment with LC water, resulting in a decrease in malondialdehyde concentration. These results suggest that LC water can reduce the SUA concentration, increase serum antioxidant activity and improve hemorheological activity in hyperuricemic rats.

3.
Cancer Sci ; 111(5): 1818-1828, 2020 May.
Article in English | MEDLINE | ID: mdl-32119176

ABSTRACT

Epstein-Barr virus (EBV) is associated with particular forms of gastric cancer (GC). We previously showed that EBV infection into gastric epithelial cells induced aberrant DNA hypermethylation in promoter regions and silencing of tumor suppressor genes. We here undertook integrated analyses of transcriptome and epigenome alteration during EBV infection in gastric cells, to investigate activation of enhancer regions and related transcription factors (TFs) that could contribute to tumorigenesis. Formaldehyde-assisted isolation of regulatory elements (FAIRE) sequencing (-seq) data revealed 19 992 open chromatin regions in putative H3K4me1+ H3K4me3- enhancers in EBV-infected MKN7 cells (MKN7_EB), with 10 260 regions showing increase of H3K27ac. Motif analysis showed candidate TFs, eg activating transcription factor 3 (ATF3), to possibly bind to these activated enhancers. ATF3 was considerably upregulated in MKN7_EB due to EBV factors including EBV-determined nuclear antigen 1 (EBNA1), EBV-encoded RNA 1, and latent membrane protein 2A. Expression of mutant EBNA1 decreased copy number of the EBV genome, resulting in relative downregulation of ATF3 expression. Epstein-Barr virus was also infected into normal gastric epithelial cells, GES1, confirming upregulation of ATF3. Chromatin immunoprecipitation-seq analysis on ATF3 binding sites and RNA-seq analysis on ATF3 knocked-down MKN7_EB revealed 96 genes targeted by ATF3-activating enhancers, which are related with cancer hallmarks, eg evading growth suppressors. These 96 ATF3 target genes were significantly upregulated in MKN7_EB compared with MKN7 and significantly downregulated when ATF3 was knocked down in EBV-positive GC cells SNU719 and NCC24. Knockdown of ATF3 in EBV-infected MKN7, SNU719, and NCC24 cells all led to significant decrease of cellular growth through an increase of apoptotic cells. These indicate that enhancer activation though ATF3 might contribute to tumorigenesis of EBV-positive GC.


Subject(s)
Activating Transcription Factor 3/metabolism , Enhancer Elements, Genetic , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/physiology , Stomach Neoplasms/genetics , Activating Transcription Factor 3/genetics , Apoptosis/genetics , Binding Sites , Cell Line , Cell Proliferation/genetics , Epigenome , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression , Herpesvirus 4, Human/genetics , Humans , Mutation , Transcriptome
4.
Oncotarget ; 9(50): 29316-29335, 2018 Jun 29.
Article in English | MEDLINE | ID: mdl-30034620

ABSTRACT

Epigenome regulates gene expression to determine cell fate, and accumulation of epigenomic aberrations leads to diseases, including cancer. NCD38 inhibits lysine-specific demethylase-1 (LSD1), a histone demethylase targeting H3K4me1 and H3K4me2, but not H3K4me3. In this study, we conjugated NCD38 with a potent small molecule called pyrrole (Py) imidazole (Im) polyamide, to analyze whether targets of the inhibitor could be regulated in a sequence-specific manner. We synthesized two conjugates using ß-Ala (ß) as a linker, i.e., NCD38-ß-ß-Py-Py-Py-Py (NCD38-ß2P4) recognizing WWWWWW sequence, and NCD38-ß-ß-Py-Im-Py-Py (NCD38-ß2PIPP) recognizing WWCGWW sequence. When RKO cells were treated with NCD38, H3K4me2 levels increased in 103 regions with significant activation of nearby genes (P = 0.03), whereas H3K4me3 levels were not obviously increased. H3K27ac levels were also increased in 458 regions with significant activation of nearby genes (P = 3 × 10-10), and these activated regions frequently included GC-rich sequences, but less frequently included AT-rich sequences (P < 1 × 10-15) or WWCGWW sequences (P = 2 × 10-13). When treated with NCD38-ß2P4, 234 regions showed increased H3K27ac levels with significant activation of nearby genes (P = 2 × 10-11), including significantly fewer GC-rich sequences (P < 1 × 10-15) and significantly more AT-rich sequences (P < 1 × 10-15) compared with NCD38 treatment. When treated with NCD38-ß2PIPP, 82 regions showed increased H3K27ac levels, including significantly fewer GC-rich sequences (P = 1 × 10-11) and fewer AT-rich sequences (P = 0.005), but significantly more WWCGWW sequences (P = 0.0001) compared with NCD38 treatment. These indicated that target regions of epigenomic inhibitors could be modified in a sequence-specific manner and that conjugation of Py-Im polyamides may be useful for this purpose.

5.
Radiat Res ; 186(1): 79-91, 2016 07.
Article in English | MEDLINE | ID: mdl-27356027

ABSTRACT

The extracellular microenvironment affects cellular responses to various stressors including radiation. Annexin A2, which was initially identified as an intracellular molecule, is also released into the extracellular environment and is known to regulate diverse cell surface events, however, the molecular mechanisms underlying its release are not well known. In this study, we found that in cultured human cancer and non-cancerous cells an extracellular release of annexin A2 was greatly enhanced 1-4 h after a single 20 cGy X-ray dose, but not after exposure to ultraviolet C (UVC) radiation. Extracellular release of annexin A2 was also enhanced after H2O2 and nicotine treatments, which was suppressed by pretreatment with the antioxidant, N-acetyl cysteine. Among the oxidative stress pathway molecules examined in HeLa cells, AMP-activated protein kinase α (AMPKα) and p38 mitogen-activated protein kinase (MAPK) were mostly activated by low-dose X-ray radiation, and the p38 MAPK inhibitor, SB203580, but not compound C (an AMPKα inhibitor), suppressed the enhancement of the annexin A2 extracellular release after low-dose X irradiation. In addition, the enhancement was suppressed in the cells in which p38α MAPK was downregulated by siRNA. HeLa cells and human cultured cells preirradiated with 20 cGy or precultured in media from low-dose X-irradiated cells showed an increase in resistance to radiation-induced cell death, and the increase was suppressed by treatment of the irradiated cell-derived media with anti-annexin A2 antibodies. In addition, extracellularly added recombinant annexin A2 conferred cellular radiation resistance. These results indicate that an oxidative stress-activated pathway via p38 MAPK was involved in the extracellular release of annexin A2, and this pathway was stimulated by low-dose X-ray irradiation. Furthermore, released annexin A2 may function in low-dose ionizing radiation-induced responses, such as radioresistance.


Subject(s)
Annexin A2/metabolism , Extracellular Space/metabolism , Extracellular Space/radiation effects , MAP Kinase Signaling System/radiation effects , Oxidative Stress/radiation effects , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Dose-Response Relationship, Radiation , Extracellular Space/drug effects , HeLa Cells , Humans , MAP Kinase Signaling System/drug effects , Oxidative Stress/drug effects , Ultraviolet Rays/adverse effects , X-Rays/adverse effects
6.
Chemistry ; 22(13): 4418-21, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26833557

ABSTRACT

A novel platinum-catalyzed cascade cyclization reaction was developed by intramolecular Friedel-Crafts-type C-H coupling of aniline derivatives with a propargyl carbonate unit-allylic amination sequence. Treatment of various propargyl carbonates tethered to meta-aniline derivatives with a Pt(dba)3/DPEphos catalyst system afforded the corresponding 3,4-fused tricyclic 3-alkylidene indolines in 42-99% yield, which were transformed into 3,4-fused indole derivatives by reaction with trifluoroacetic acid. The reaction products exhibited antiproliferative activities against cancer cells, but not normal cells, revealing the potential usefulness of this reaction for medicinal chemistry.


Subject(s)
Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/chemical synthesis , Indoles/chemistry , Indoles/chemical synthesis , Platinum/chemistry , Amination , Catalysis , Molecular Structure , Stereoisomerism
7.
ACS Omega ; 1(6): 1164-1172, 2016 Dec 31.
Article in English | MEDLINE | ID: mdl-30023504

ABSTRACT

Aberrant DNA methylation causes major epigenetic changes and has been implicated in cancer following the inactivation of tumor suppressor genes by hypermethylation of promoter CpG islands. Although methylated DNA regions can be randomly demethylated by 5-azacytidine and 5-aza-2'-deoxycytidine, site-specific inhibition of DNA methylation, for example, in the promoter region of a specific gene, has yet to be technically achieved. Hairpin pyrrole (Py)-imidazole (Im) polyamides are small molecules that can be designed to recognize and bind to particular DNA sequences. In this study, we synthesized the hairpin polyamide MLH1_-16 (Py-Im-ß-Im-Im-Py-γ-Im-Py-ß-Im-Py-Py) to target a CpG site 16 bp upstream of the transcription start site of the human MLH1 gene. MLH1 is known to be frequently silenced by promoter hypermethylation, causing microsatellite instability and a hypermutation phenotype in cancer. We show that MLH1_-16 binds to the target site and that CpG methylation around the binding site is selectively inhibited in vitro. MLH1_non, which does not have a recognition site in the MLH1 promoter, neither binds to the sequence nor inhibits DNA methylation in the region. When MLH1_-16 was used to treat RKO human colorectal cancer cells in a remethylating system involving the MLH1 promoter under hypoxic conditions (1% O2), methylation of the MLH1 promoter was inhibited in the region surrounding the compound binding site. Silencing of the MLH1 expression was also inhibited. Promoter methylation and silencing of MLH1 were not inhibited when MLH1_non was added. These results indicate that Py-Im polyamides can act as sequence-specific antagonists of CpG methylation in living cells.

8.
Int J Oncol ; 47(5): 1839-44, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26398575

ABSTRACT

There is an ongoing search for plant-derived sesquiterpenes, particularly for those with anticancer activity against human cancer cells. The sesquiterpene ineupatorolide B (InB), isolated from the Inula cappa, showed potent growth-inhibitory activity against HeLa cells but less activity against MM1-CB melanoma cells. Staining by terminal deoxynucleotidyl transferase dUTP nick-end labeling method revealed that this activity was, at least in part, due to the induction of apoptosis. The activities of major transcription factors were examined by using a luciferase reporter assay. The results showed that the transactivation ability of nuclear factor of activated T-cell (NFAT) was enhanced. The activation of NFAT by InB was largely suppressed by preincubation with protein kinase C (PKC) inhibitors such as staurosporine and K252a. Western blot analysis revealed that the the levels of phosphorylated PKCα, but not other subtypes, increased after treatment with InB. Knockdown of PKCα using siRNA attenuated the cytotoxic activity of InB. Thus, InB may exhibit growth-inhibitory activity through the activation of PKCα, followed by an increase in NFAT transactivation ability.


Subject(s)
NFATC Transcription Factors/genetics , Protein Kinase C-alpha/biosynthesis , Sesquiterpenes/administration & dosage , Uterine Cervical Neoplasms/genetics , Apoptosis/drug effects , Female , HeLa Cells , Humans , Inula/chemistry , NFATC Transcription Factors/biosynthesis , Phosphorylation , Protein Kinase C-alpha/genetics , RNA, Small Interfering , Uterine Cervical Neoplasms/pathology
9.
No To Hattatsu ; 47(4): 298-303, 2015 Jul.
Article in Japanese | MEDLINE | ID: mdl-26353453

ABSTRACT

OBJECTIVE: To investigate the protective function of Cockayne syndrome (CS) patient-derived cells against oxidative stress, we examined the sensitivity to cell death and the repair activity of DNA damages after exposure to oxidative stress in CS cells. METHODS: We used two CS cell lines, CS3BES (CSA defective) and CSIANS (CSB defective), the human cervical cancer cell line HeLa cells, and the human fibroblastic cell line RSa. Cells were exposed to oxidative stresses, such as X-ray irradiation and hydrogen peroxide treatment, and the sensitivity to cell death was examined using the colony survival assay and MTT assay. DNA lesions were analyzed using the comet assay. RESULTS: CS3BES and CS1ANS cells showed higher sensitivity to cell death induced by X ray and hydrogen peroxide than HeLa and RSa cells. Furthermore, after exposure to the stresses the levels of DNA damage were higher, or repair activity was lower in CS3BES cells when compared with HeLa cells. CONCLUSIONS: The present results clearly show that the two CS cell lines are vulnerable to oxidative stress and suggest that both CSA and CSB proteins are involved in the protective response against oxidative injury.


Subject(s)
Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , DNA Damage , DNA Repair , Cell Death/drug effects , Cell Death/radiation effects , Cell Line , Humans , Hydrogen Peroxide/pharmacology , Oxidative Stress
10.
Mol Med Rep ; 12(4): 6215-20, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26239548

ABSTRACT

Hochu­ekki­to (HET), a Kampo herbal medicine composed of ten medicinal plants, is traditionally used to improve the general state of patients with malignant diseases such as cancer. Recent studies showed that HET had an anti­cancer effect against several cancer cell lines in vitro by inducing apoptosis. However, high doses of HET may have cytotoxic effects attributed to saponins or detergent­like compounds. Therefore, the present study used low doses of HET (50 µg/ml), which did not affect cell viability, to evaluate its synergistic anti­cancer effects with cisplatin. HeLa cells were cultured for 24 h with 50 µg/ml HET, followed by cisplatin treatment for 24 h at various concentrations. Subsequently, the sensitivity of the cells to cisplatin was assessed using a colony survival and a crystal violet cell viability assay. Furthermore, cisplatin­induced apoptosis was analyzed by flow cytometry. Proteins associated with cell viability and apoptosis, including phosphorylated (p­)Akt, p53, B­cell lymphoma 2 (Bcl­2), Bcl­2­associated X protein (Bax) and active caspase­3 were analyzed by immunoblotting. The present study revealed that cell survival was decreased and apoptosis was increased in HeLa cells pre­treated with HET prior to cisplatin treatment compared with HET­untreated cells. Furthermore, protein expression of p53 and active caspase­3 was increased, while the expression of p­Akt as well as the Bcl­2/Bax ratio, an index of survival activity in cells, were decreased in the HET­pre­treated cells compared with those in HET­untreated cells following incubation with cisplatin. In conclusion, the present study indicated that HET enhanced cisplatin­induced apoptosis of HeLa cells and that the administration of HET may therefore be clinically beneficial alongside apoptosis­inducing chemotherapy.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , Drugs, Chinese Herbal/pharmacology , Antineoplastic Agents/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , Cell Survival/drug effects , HeLa Cells , Humans , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
11.
Int J Oncol ; 44(5): 1685-90, 2014 May.
Article in English | MEDLINE | ID: mdl-24584142

ABSTRACT

miRNAs are small non-coding RNAs that inhibit gene expression by cleaving or hindering the translation of target mRNAs. In this study, we focused on miR-431, which mediated inhibition of cell viability by human interferon-ß (HuIFN-ß). We aimed to demonstrate an antineoplastic effect of HuIFN-ß via miR-431 expression against medulloblastoma and glioblastoma, because HuIFN-ß is frequently used in adjuvant therapy of these tumors. Addition of HuIFN-ß to medulloblastoma and glioblastoma cells reduced viability, significantly decreased miR-431 expression, upregulated expression of SOCS6 (putative miR-431 target genes) and inhibited Janus kinase (JAK) 1 and signal transducer and activator of transcription (STAT) 2. The mitogen-activated protein kinase (MAPK) pathway, but not the phosphoinositide 3-kinase (PI3K)-Akt pathway, was downregulated in medulloblastoma cells, whereas the PI3K-Akt pathway, but not the MAPK pathway, was downregulated in glioblastoma cells. Addition of HuIFN-ß and transient transfection with miR-431 to medulloblastoma and glioblastoma cells did not reduce viability, downregulated expression of SOCS6, and concomitantly activated the JAK1 and STAT2. We propose that, in medulloblastoma and glioblastoma cells, HuIFN-ß decreases miR-431 expression and upregulates SOCS6 expression, and consequently inhibit cell proliferation by suppressing the JAK-STAT signaling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Down-Regulation/drug effects , Glioma/metabolism , Interferon-beta/pharmacology , MicroRNAs/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioma/pathology , Humans , Signal Transduction/drug effects , Suppressor of Cytokine Signaling Proteins/metabolism
12.
J Gastroenterol Hepatol ; 29(4): 736-41, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24224878

ABSTRACT

BACKGROUND AND AIM: Gastric ulcer healing is a complex process involving cell proliferation and tissue remodeling. Sonic hedgehog (Shh) activates the Shh signaling pathway, which plays a key role in processes such as tissue repair. Shh and interleukin 1ß (IL1ß) have been reported to influence the proliferation of gastric mucosa. We evaluated the relationships between the speed of gastric ulcer healing and the levels of expression of Shh and IL1ß. METHODS: The study included 45 patients (mean age 71.9 ± 9.0 years; M/F, 30/15) who underwent endoscopic submucosal dissection (ESD) for gastric cancer, followed by standard dose of oral proton-pump inhibitor for 4 weeks. Subsequently, the size of ESD-induced artificial ulcers were measured to determine the speed of gastric ulcer healing, and regenerating mucosa around the ulcers and appropriately matched controls were collected from patients by endoscopic biopsy. Polymerase chain reaction (PCR) array analysis of genes in the Shh signaling pathway was performed, and quantitative reverse transcription (RT)-PCR was used to measure IL1ß mRNA. RESULTS: The levels of Shh and IL1ß mRNA were 3.0 ± 2.7-fold and 2.5 ± 2.5-fold higher, respectively, in regenerating mucosa of artificial ulcers than in appropriately matched controls, with the two being positively correlated (r = 0.9, P < 0.001). Shh (r = 0.8, P < 0.001) and IL1ß (r = 0.7, P < 0.005) expression was each positively correlated with the speed of gastric ulcer healing, but multivariate analysis showed that Shh expression was the only significant parameter (P = 0.045). CONCLUSIONS: Expression of Shh was correlated with the speed of gastric ulcer healing, promoting the regeneration of gastric mucosa.


Subject(s)
Gastric Mucosa/physiology , Gene Expression Regulation, Developmental/genetics , Hedgehog Proteins/physiology , Regeneration/genetics , Signal Transduction , Stomach Ulcer/physiopathology , Wound Healing/genetics , Aged , Aged, 80 and over , Female , Hedgehog Proteins/genetics , Humans , Interleukin-1beta/genetics , Interleukin-1beta/physiology , Male , Middle Aged , Multivariate Analysis , RNA, Messenger/analysis , Time Factors , Wound Healing/physiology
13.
Biosci Biotechnol Biochem ; 77(2): 307-11, 2013.
Article in English | MEDLINE | ID: mdl-23391921

ABSTRACT

Annexin II, an HSP27-interacting protein, is involved in the protection of human cells against UVC. UVB is concerned with deleterious actions on human health. In this study, we attempted to confirm the anti-UVB effect of annexin II, and to elucidate the mechanisms underlying annexin II-involving UV resistance. The RSa cells were more sensitive to UVB lethality than the AP(r)-1 cells. Overproduction of annexin II in RSa cells resulted in increased resistance to UVB lethality, while annexin II siRNA-transfected AP(r)-1 cells were sensitized to UVB lethality. The excision capacity of the two major types (CPD and 6-4PP) of UVC- and UVB-damaged DNA in AP(r)-1 cells was greater than in RSa cells. The excision capacity of the RSa cells improved following upregulation of annexin II, while the capacity of the AP(r)-1 cells decreased after annexin II downregulation. Our results suggest that annexin II is involved in the UV resistance of human cells, via functioning in nucleotide excision repair.


Subject(s)
Annexin A2/genetics , DNA Repair/radiation effects , DNA/genetics , Fibroblasts/radiation effects , Radiation Tolerance/genetics , Annexin A2/antagonists & inhibitors , Annexin A2/metabolism , Cell Death/radiation effects , Cell Line , Cell Proliferation/radiation effects , DNA/chemistry , DNA Damage , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation/radiation effects , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Molecular Chaperones , Organ Specificity , RNA, Small Interfering/genetics , Signal Transduction/radiation effects , Ultraviolet Rays
14.
Exp Ther Med ; 4(5): 913-917, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23226748

ABSTRACT

Revealing the key molecules regulating the stress-response pathways in human cells is an intriguing problem. Chaperones, such as glucose-regulated protein 78 (GRP78) and heat shock protein 27 (HSP27), are important molecules for protecting the viability of human cells; however, it remains to be further clarified whether the molecules differentially modulate cellular responses to various types of stressors, such as DNA-damaging ultraviolet ray C (principally 254-nm wavelength, UVC) and cytocidal cytokine interferons. In the present study, the human breast cancer cell lines KT and MCF-7 were examined for GRP78 and HSP27 expression following exposure to UVC and human interferon-ß (HuIFN-ß). The KT cells demonstrated a higher sensitivity to both UVC and HuIFN-ß lethality than MCF-7 cells. The cellular expression levels of GRP78 in KT cells, assessed by western blot analysis, were approximately 2-fold higher than that in MCF-7 cells, while the expression of HSP27 in the KT cells was 20% of the expression in the MCF-7 cells. Decreased resistance to UVC lethality was observed in GRP78 siRNA-transfected KT cells. In addition, HSP27 cDNA transfection of KT cells resulted in an increased resistance to UVC lethality. The cDNA-transfected KT cells showed an increased viability against HuIFN-ß, compared with that of empty vector-transfected cells. By contrast, KT cells pretreated with HuIFN-ß and irradiated with UVC demonstrated an increased resistance to UVC lethality, in association with increased levels of HSP27 expression. Thus, HSP27 may control the survival response pathways to both UVC and HuIFN-ß in the human cells examined.

15.
Pancreas ; 41(8): 1247-54, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22750966

ABSTRACT

OBJECTIVES: Extracellular microenvironment plays crucial roles in the development of cancers and chemoresistance. Pancreatic carcinoma is resistant to almost all chemotherapeutic agents. In this study, we identified annexin II in the medium from pancreatic cancer cells as a protein released into the extracellular environment. METHODS: Medium from 5-hour cultures of various cancer cells was collected. Proteins in the medium were detected by molecular mass analysis and immunoblotting. Anticancer drug sensitivity of cells preincubated with or without recombinant annexin II (rANX II) was measured using crystal violet assay and colony survival assay. Apoptosis-related molecules were analyzed by immunoblotting. RESULTS: Recombinant ANX II supplementation in the medium confers resistance to anticancer drugs, including cisplatin, 5-fluorouracil, and gemcitabine, in MiaPaCa-2 and AsPC-1 cells. In MiaPaCa-2 cells, rANX II supplementation resulted in suppression of caspase-3 activation associated with increased Bcl-2/Bax ratios. Suppression of cisplatin-induced cell death by rANX II supplementation was canceled by inhibitors of phosphatidylinositol 3-kinase and mitogen-activated protein kinase kinase signal pathways. CONCLUSIONS: The current study is the first report to demonstrate that supplementation of rANX II in the medium increased resistance to anticancer drugs in pancreatic cancer cells. Recombinant ANX II exerts cell death-suppressive function by antagonizing cisplatin-induced apoptosis.


Subject(s)
Annexin A2/pharmacology , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Pancreatic Neoplasms/drug therapy , Annexin A2/metabolism , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Humans , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism , Pancreatic Neoplasms
16.
Int J Oncol ; 40(5): 1470-6, 2012 May.
Article in English | MEDLINE | ID: mdl-22293894

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that inhibit gene expression by cleaving or hindering the translation of target mRNAs. We used microarray-based comparative transcriptome analysis to identify changes in miRNA expression and function between a human cell line, RSa, which is highly sensitive to HuIFN-ß-mediated inhibition of cell viability, and its variant, the F-IFr cell line, which is relatively resistant to the cytokine. miR-431 expression was significantly higher in RSa cells compared with F-IFr cells. The addition of HuIFN-ß to RSa cultures reduced cell viability, down-regulated expression of IGF1R and IRS2 (putative miR-431 target genes), and inhibited the PI3K-Akt and MAPK pathways. The survival of F-IFr cells was not reduced by HuIFN-ß, but transient transfection with miR-431 precursors significantly decreased viability and concomitantly down-regulated IGF1R and IRS2 expression. In addition, the MAPK pathway, but not the PI3K-Akt pathway, was suppressed in F-IFr cells. Based on these results, we propose that, in RSa cells, HuIFN-ß-induced miR-431 expression may down-regulate IGF1R and IRS2 expression, and consequently inhibit cell proliferation by suppressing the MAPK pathway.


Subject(s)
Fibroblasts/metabolism , Interferon-beta/metabolism , MicroRNAs/metabolism , Cell Line, Transformed , Cell Proliferation , Cell Survival , Down-Regulation , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/pathology , Gene Expression Profiling/methods , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , MAP Kinase Signaling System , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Time Factors , Transfection , Up-Regulation
17.
Radiat Res ; 176(6): 732-42, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22141411

ABSTRACT

In this study, we found that refractoriness to ultraviolet (UVC) light-induced cell death was increased in UVC-radiation-sensitive cells derived from Cockayne syndrome patients when the cells were precultured in medium supplemented with recombinant annexin II (rANX II). In CS3BES cells, an immortal cell line derived from Cockayne syndrome patients, the rANX II supplementation-induced UVC-radiation resistance was suppressed by treatment with an anti-annexin II antibody and EGTA. The amount of biotinylated annexin II on the cell surface increased in the rANX II-supplemented cells but did not increase in the cells that were cotreated with rANX II and EGTA. The capacity to remove UVC-radiation-damaged DNA, (6-4) photoproducts and cyclobutane pyrimidine dimers, was the same in cells that were precultured with rANX II and in control cells that did not receive rANX II supplementation. The rANX II supplementation-induced UVC-radiation resistance was also observed in nucleotide excision repair-deficient cells and xeroderma pigmentosum group A-downregulated cells. The Bcl-xL to Bax protein ratios, an index of survival activity in cells exposed to lethal stresses, were increased in the cells that had been precultured in rANX II for 24 h prior to UVC irradiation. Treatment with a phosphatidylinositol 3-kinase inhibitor suppressed the increased UVC-radiation resistance and Bcl-xL to Bax ratios in the cells with rANX II supplementation. Furthermore, downregulation of Bcl-xL by siRNA transfection also suppressed the UVC-radiation resistance that was induced by rANX II supplementation. These results suggest that the increase in the Bcl-xL to Bax ratios may be associated with enhanced resistance to UVC-radiation-induced cell death.


Subject(s)
Annexin A2/pharmacology , Extracellular Space , Radiation Tolerance/drug effects , Recombinant Proteins/pharmacology , Ultraviolet Rays , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Cell Culture Techniques , Cell Survival/drug effects , Cell Survival/radiation effects , Cockayne Syndrome/pathology , Down-Regulation/drug effects , Down-Regulation/radiation effects , Enzyme Inhibitors/pharmacology , Extracellular Space/drug effects , Extracellular Space/radiation effects , HeLa Cells , Humans , Phosphoinositide-3 Kinase Inhibitors , Radiation Tolerance/radiation effects
18.
Biosci Biotechnol Biochem ; 75(9): 1685-91, 2011.
Article in English | MEDLINE | ID: mdl-21897041

ABSTRACT

Little is known about the ability of miso, to modulate mutability in human cells. We have observed increased levels of glucose-regulated protein 78 (GRP78) expression in association with suppression of mutation in human RSa cells irradiated with ultraviolet C (UVC). Here we examined to determine whether miso treatment results in increased GRP78 expression and suppression of UVC mutagenicity in RSa cells. Supernatants of water extracts of miso products and their components were tested. In the sample-treated cells, the amount of GRP78, as estimated by RT-PCR and immunoblotting analysis, increased, and the UVC-induced ouabain resistant mutation (Oua(R)) and the K-ras codon 12-base substitution mutation frequency decreased. This decrease was not observed in cells with downregulation of GRP78 by GRP78 siRNA transfection. The results suggest that miso suppresses UVC mutagenicity by increasing GRP78 expression in human cells.


Subject(s)
Fibroblasts/drug effects , Fibroblasts/radiation effects , Gene Expression , Heat-Shock Proteins , Mutation , Plant Extracts/pharmacology , Soy Foods , Blotting, Western , Cell Line, Transformed , Codon , Endoplasmic Reticulum Chaperone BiP , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression/drug effects , Gene Expression/radiation effects , Gene Silencing/drug effects , Genes, ras/drug effects , Genes, ras/radiation effects , Heat-Shock Proteins/agonists , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Japan , Mutation/drug effects , Mutation/radiation effects , Mutation Rate , Ouabain/pharmacology , Plant Extracts/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Ultraviolet Rays/adverse effects
19.
Oncol Rep ; 25(3): 879-85, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21225231

ABSTRACT

Glucose-regulated protein 78 (GRP78) is expressed abundantly in various types of cancer cells and is believed to contribute to chemotherapeutic resistance. In this study, we investigated the effect of a continuous approach for the expression of a short hairpin RNA (shRNA) targeted to GRP78 with retrovirus transduction on the sensitivity to the anticancer drugs VP-16 and cisplatin. The reduction of GRP78 expression failed, and the expression of GRP94 and P5 chaperon mRNA increased; this increase was associated with a mild activation of the unfolded protein response in HeLa cells, which were stably transduced with GRP78 shRNA gene. The transduced cells exhibited similar sensitivity to VP-16-induced cell death when compared to control GFP shRNA gene-transduced cells. However, sensitivity to cisplatin-induced cell death was higher in GRP78 shRNA gene-transduced cells compared to control cells. These results demonstrate that the continuous or prolonged approach targeting GRP78 confers sensitization of HeLa cells to cisplatin independently of the down-regulation of GRP78 expression. The role of the unfolded protein response in sensitization to cisplatin is discussed.


Subject(s)
Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Genetic Therapy/methods , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Neoplasms/therapy , RNA, Small Interfering/genetics , Antineoplastic Agents/therapeutic use , Base Sequence , Cells, Cultured , Combined Modality Therapy , Down-Regulation/genetics , Drug Resistance, Neoplasm/drug effects , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation, Neoplastic/genetics , Genetic Vectors , HeLa Cells , Humans , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Retroviridae/genetics , Retroviridae/physiology
20.
Mol Med Rep ; 3(4): 673-8, 2010.
Article in English | MEDLINE | ID: mdl-21472297

ABSTRACT

Two abietane and one labdane type diterpenoids were isolated from the methanol extracts of Torreya nucifera pulp and investigated for their ability to inhibit the growth of human cancer cells. Among the three compounds, the labdane compound kayadiol was found to have the most effective inhibitory effect against a wide variety of human cancer cells. Using the MTT assay, kayadiol was determined to have an IC50 (50% inhibition concentration) of 30 µM in HeLa cells, and also to exhibit anti-proliferative effects towards six other human cancer cell lines, with IC50 values of 30-50 µM. Kayadiol treatment of HeLa cells resulted in a dose-dependent generation of apoptotic events, including DNA laddering (≤100 µM). Moreover, kayadiol-treated HeLa cells showed activation of caspases-3 and -9, as well as an increase in the depolarization of mitochondrial membrane potential and the Bax/Bcl-2 ratio. These results indicate that a mitochondria-related apoptotic pathway is involved in the kayadiol-induced death of HeLa cells. Kayadiol is therefore a promising novel anti-proliferative agent and merits further investigation.

SELECTION OF CITATIONS
SEARCH DETAIL
...