Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
In Vivo ; 34(6): 3195-3203, 2020.
Article in English | MEDLINE | ID: mdl-33144424

ABSTRACT

BACKGROUND/AIM: Gemcitabine, an inhibitor of DNA synthesis, is the gold standard chemotherapeutic agent for pancreatic ductal adenocarcinoma (PDAC). MicroRNAs (miRNAs) play critical roles in cancers, including PDAC. However, less is known about the effect of gemcitabine on PDAC cells and miRNA expression in PDAC. We evaluated the effect of gemcitabine on the cell cycle of PDAC cells in vitro and in vivo and on the miRNA expression profile. MATERIALS AND METHODS: Effects of gemcitabine on PK-1 and PK-9 cell growth were evaluated using a cell counting kit-8 assay. Xenografted mouse models were used to assess gemcitabine effects in vivo. RESULTS: Gemcitabine inhibited the proliferation and tumour growth of PK-1 cells, and induced S phase cell cycle arrest. Numerous miRNAs were altered upon gemcitabine treatment of PK-1 cells and xenograft models. CONCLUSION: Altered miRNAs may serve as potential therapeutic targets for improving the efficacy of gemcitabine in PDAC.


Subject(s)
MicroRNAs , Pancreatic Neoplasms , Animals , Apoptosis , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Deoxycytidine/analogs & derivatives , Mice , MicroRNAs/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Xenograft Model Antitumor Assays , Gemcitabine
2.
Oncol Lett ; 20(2): 1727-1733, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32724415

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Clinical management has improved the prognosis of early HCC, but that of advanced HCC remains poor. Sorafenib, an oral multikinase inhibitor, provided a treatment option for advanced-stage HCC, and prolonged the survival and inhibited tumor progression as first-line therapy in patients with advanced HCC. In this study, we investigated if specific microRNAs could act as predictive biomarkers of sorafenib effectiveness and indicate the best time to switch to second-line therapies. Sorafenib inhibited the proliferation of the Li-7, Hep3B, HepG2 and Huh7 liver cancer cell lines (effective group), but not that of the HLE, HLF and ALEX cancer cell lines (non-effective group). A microRNA (miRNA/miR) analysis was performed comparing sorafenib-effective and non-effective cells lines as well as serum samples from patients with HCC from sorafenib-effective (complete response/partial response) and -non-effective (progressive disease) groups before sorafenib administration and detected three differentially-expressed miRNAs that were common among the in vivo and in vitro samples. The increase rate (effective/non-effective) of hsa-miR-30d in the medium was higher than that in the cancer cells. hsa-miR-30d was highly expressed in the serum and exosomes of patients with HCC in the effective group when compared to those of the non-effective group. Additionally, the hsa-miR-30d expression in the medium of cancer cell lines was highly upregulated in the effective group compared with the non-effective group. These results suggested that hsa-miR-30d might be secreted by the cancer cells to the serum through the exosomes. We identified a specific circulating miRNA that is related to refractory HCC under sorafenib therapy. Therefore, hsa-miR-30d might serve as a predictive biomarker for the efficacy of sorafenib therapy in HCC.

3.
Oncol Rep ; 44(1): 339-348, 2020 07.
Article in English | MEDLINE | ID: mdl-32627043

ABSTRACT

Gastric cancer is one of the most common malignancies diagnosed worldwide. Telmisartan, an angiotensin receptor blocker (ARB), suppresses the proliferation of cancer cells and the growth of tumors through an unknown mechanism. To identify the mechanism, the present study was designed to evaluate the effects of telmisartan on gastric cancer cell lines and tumors in vitro and in vivo and the associated signaling molecules were identified. It was shown here that telmisartan suppressed the proliferation of the cultured human gastric cancer cell lines MKN74, MKN1 and MKN45 as detected in the CCK­8 assay. In a mouse xenograft model of gastric cancer, telmisartan suppressed tumor growth by arresting the cell cycle at the G0/G1 phase through inhibition of the expression of cyclin D1, the catalytic subunit of cyclin dependent kinase 4 (CDK4), as well as the phosphorylation of the tumor suppressor retinoblastoma (pRb) protein as detected by western blotting. Notably, telmisartan did not induce apoptosis, as indicated by consistent levels of caspase­cleaved keratin 18 in MKN74 cells. Furthermore, telmisartan inhibited the phosphorylation of epidermal growth factor receptor (EGFR) and increased the levels of the angiogenesis­related protein tissue inhibitor of metalloproteinase­1 (TIMP­1). Analyses of microarrays revealed that telmisartan altered the expression of miRNAs in MKN74 cells. In conclusion, telmisartan suppressed the proliferation of human gastric cancer cells by inducing cell cycle arrest.


Subject(s)
Cyclin D1/genetics , Cyclin-Dependent Kinase 4/genetics , Retinoblastoma Protein/metabolism , Stomach Neoplasms/drug therapy , Telmisartan/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Repositioning , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , MicroRNAs/genetics , Phosphorylation/drug effects , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Telmisartan/pharmacology , Xenograft Model Antitumor Assays
4.
Anticancer Res ; 40(1): 121-132, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31892560

ABSTRACT

BACKGROUND/AIM: Pancreatic neuroendocrine tumors (pNETs) are rare pancreatic neoplasms, and therapeutic options for pNETs are limited. Metformin is an anti-hypoglycemic drug that appears to have anticancer effects. However, little is known about the effect of metformin on pNETs. In this study, we investigated the anti-proliferative effect of metformin on a human pNET cell line. MATERIALS AND METHODS: The anti-proliferative properties of metformin were evaluated in QGP-1 and NCI-H727 cells using a cell counting kit-8 assay. Xenograft mouse models were used to assess the tumor effect in vivo. RESULTS: Metformin inhibited the proliferation and anti-tumor growth of QGP-1 cells, accompanied by their arrest during the cell cycle at the G0/G1 phase. Immunohistochemical analysis of tumor tissues revealed down-regulation of cyclin D1 and proliferating cell nuclear antigen in the metformin-treated group. Additionally, metformin induced apoptosis, and the expression of survivin and claspin were decreased in metformin-treated QGP-1 cells according to the apoptosis array. Furthermore, the angiogenic related protein TIMP-1 was down-regulated, and its miRNA expression was altered by metformin in QGP-1 cells. CONCLUSION: Taken together, our study demonstrated the therapeutic potential of metformin and provides molecular mechanistic insights into its anti-tumoral effect on pNETs. This study is the first one describing anti-tumoral effects in pNETs.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Metformin/pharmacology , Biomarkers , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , MicroRNAs/genetics , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
5.
Disabil Rehabil Assist Technol ; 15(5): 528-536, 2020 07.
Article in English | MEDLINE | ID: mdl-31012763

ABSTRACT

Purpose: In patients with mild cognitive impairment and Alzheimer's disease (AD), faculties associated with instrumental activities of daily living (IADL) decline owing to reduced cognitive function. One type of IADL is medication behaviour. Medication management is critical for patients with AD. Previous studies have reported that presentations using speech and images are effective for facilitating IADL autonomy but there are few examinations of effective presentation methods. Therefore, we examined what kind of display methods are effective in helping patients with AD with medication management.Materials and methods: Ten healthy elderly and 9 patients with mild AD were asked to perform the task of taking out medicine bags from a case at a designated time. We gave 3 kinds of instructions and examined the differences in participants' reaction times. Task 1 included verbal instructions alone, Task 2 included verbal instructions and pictorial and written instructions, and Task 3 used a video conference system (presenting pictorial, written, and verbal instructions) at a designated time. Task 3 could be conducted remotely over the internet. The relationship between these results and neuropsychological tests was also explored.Results: Task 3 was an effective method for patients with mild AD. In addition, we found correlations between the methods of Tasks 1-3 and the Japanese version of the Mini-Mental State Examination (MMSE-J).Conclusions: The method of Task 3 may lead to home support for patients with AD. The MMSE-J could be used to identify changes in the adaptive functioning of patients exposed to distinct presentation methods.Implications for rehabilitationAppropriate transmission methods will increase the IADL autonomy of patients with mild AD who have memory impairment. While still images alone are not effective for patients with mild AD, when combined with verbal instructions, they prove effective for this group.The results of this study are useful for providing patients with mild AD with support in their IADLs, especially when methods that use images plus speech are employed. As participants were instructed via the internet, this study shows a way to help patients with mild AD even from a remote location.Until now, no studies have examined the adaptation criteria for instructive methods for patients with mild AD. This study shows that the MMSE could be used to determine the applicability of these instructive methods. The identification of cut-off values in future research could lead to more effective IADL support.


Subject(s)
Alzheimer Disease/psychology , Cognitive Dysfunction/psychology , Computer User Training , Medication Therapy Management , Activities of Daily Living , Aged , Aged, 80 and over , Alzheimer Disease/drug therapy , Cognitive Dysfunction/drug therapy , Female , Humans , Male , Neuropsychological Tests , Task Performance and Analysis
6.
Anticancer Drugs ; 31(3): 231-240, 2020 03.
Article in English | MEDLINE | ID: mdl-31815765

ABSTRACT

Gallbladder cancer is the most common biliary tract cancer with poor prognosis and wide variation in incidence rates worldwide, being very high in some countries in Latin America and Asia. Treatment of type 2 diabetes with metformin causes a reduction in the incidence of cancer. Till date, there are no reports on the anti-tumor effects of metformin in gall bladder cancer. Therefore, this study evaluated the effects of metformin on the proliferation of human gallbladder adenocarcinoma cells in vitro and in vivo, as well as explored the microRNAs associated with the anti-tumor effects of metformin. Metformin inhibited the proliferation in gallbladder adenocarcinoma cell lines NOZ, TGBC14TKB, and TGBC24TKB, and blocked the G0 to G1 transition in the cell cycle. This was accompanied by strong reduction in the expression of G1 cyclins, especially cyclin D1 and its catalytic subunits including cyclin-dependent kinase 4, and in retinoblastoma protein phosphorylation. In addition, metformin reduced the phosphorylation of receptor tyrosine kinases, especially Tie-2, ALK, PYK, EphA4, and EphA10, as well as angiogenesis-related proteins, including RANTES, TGF-ß, and TIMP-1. Moreover, metformin also markedly altered microRNA expression profile leading to an anti-tumor effect. Treatment of athymic nude mice bearing xenograft tumors with metformin inhibited tumor growth. These results suggest that metformin may be used clinically for the treatment of gallbladder adenocarcinoma.


Subject(s)
G1 Phase Cell Cycle Checkpoints/drug effects , Gallbladder Neoplasms/drug therapy , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Resting Phase, Cell Cycle/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Gallbladder Neoplasms/pathology , Humans , Male , Metformin/therapeutic use , Mice , Mice, Inbred BALB C , MicroRNAs , Neovascularization, Physiologic/drug effects , Receptor Protein-Tyrosine Kinases/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
8.
Can J Gastroenterol Hepatol ; 2018: 3983707, 2018.
Article in English | MEDLINE | ID: mdl-29850454

ABSTRACT

Background: Endoscopic transpapillary gallbladder stenting (ETGBS) is an effective procedure for treating high-risk patients with acute cholecystitis and severe comorbidities. However, the efficacy of ETGBS for recurrent cholecystitis (RC) remains unclear. This study aimed to explore its efficacy in patients with RC for whom cholecystectomy is contraindicated because of its high surgical risk. Methods: Data on 19 high-risk patients who had undergone ETGBS for RC after initial conservative therapy in our institution between June 2006 and May 2012 were retrospectively examined. The primary outcome was the clinical success rate, which was defined as no recurrences of acute cholecystitis after ETGBS until death or the end of the follow-up period. Secondary outcomes were technical success rate and adverse events (AEs). Results: The clinical success rate of ETGBS was 100%, the technical success rate 94.7%, and AE rate 5%: one patient developed procedure-related mild acute pancreatitis. The clinical courses of all patients were as follows: four died of nonbiliary disease, and the remaining 15 were subsequently treated conservatively. The median duration of follow-up was 14.95 months (range 3-42 months). Conclusions: ETGBS is an effective alternative for managing RC in high-risk patients with severe comorbidities.


Subject(s)
Cholangiopancreatography, Endoscopic Retrograde/methods , Cholecystitis, Acute/therapy , Stents , Aged , Aged, 80 and over , Ampulla of Vater , Cholangiopancreatography, Endoscopic Retrograde/adverse effects , Conservative Treatment , Female , Gallbladder , Humans , Male , Middle Aged , Recurrence , Retreatment , Retrospective Studies , Stents/adverse effects , Time Factors
9.
PLoS One ; 13(5): e0198142, 2018.
Article in English | MEDLINE | ID: mdl-29795674

ABSTRACT

MicroRNAs (miRNAs) are short, endogenous RNAs that post-transcriptionally repress mRNAs. Over the course of evolution, many new miRNAs are known to have emerged and added to the existing miRNA repertoires of drosophilids and vertebrates. Despite the large number of miRNAs in existence, the complementary pairing of only ~7 bases between miRNAs and mRNAs is sufficient to induce repression. Thus, miRNA targeting is so widespread that genes coexpressed with a miRNA have evolved to avoid sites that are targeted by the miRNA. Besides this avoidance, little is known about the preferential modes of miRNA targeting. Therefore, to elucidate miRNA targeting preference and avoidance, we evaluated the bias of the number of miRNA targeting occurrences in relation to expression intensities of miRNAs and their coexpressed target mRNAs by surveying transcriptome data from human organs. We found that miRNAs preferentially target genes with intermediate levels of expression, while avoiding highly expressed ones, and that older miRNAs have greater targeting specificity, suggesting that specificity increases during the course of evolution.


Subject(s)
Evolution, Molecular , Gene Expression Regulation , Mammals/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome , Animals , Humans
10.
Oncol Lett ; 15(1): 407-414, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29387226

ABSTRACT

Pancreatic cancer is the eighth-leading cause of cancer-associated mortality in males and the ninth-leading cause in females worldwide. Even when diagnosed early enough to be potentially resectable, the prognosis of invasive pancreatic cancer is poor. Galectin-9 (Gal-9) is a tandem-repeat type galectin that has recently been demonstrated to possess an anti-proliferative effect on cancer cells. Therefore, the present study evaluated the effects of Gal-9 on the proliferation of human pancreatic cancer cells and examined the microRNAs that are associated with the antitumor effects of Gal-9. Gal-9 suppressed the proliferation of multiple pancreatic cancer cell lines. In addition, Gal-9 treatment increased the levels of caspase-cleaved keratin 18 and the expression of cytochrome c in pancreatic cancer cell lines. This data suggests that Gal-9 induces intrinsic apoptosis in pancreatic cancer cell lines through the caspase-dependent and caspase-independent pathways. In addition, Gal-9 reduced the expression levels of phosphorylated epidermal growth factor receptor and numerous receptor tyrosine kinases (RTKs). In conclusion, Gal-9 may suppress the growth of human pancreatic cancer cells in vitro. These findings suggest that Gal-9 may be a new therapeutic agent for the treatment of pancreatic cancer.

11.
Endosc Int Open ; 5(8): E695-E705, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28782002

ABSTRACT

BACKGROUND AND STUDY AIMS: Endoscopic ultrasound-guided fine needle aspiration (FNA) for gastrointestinal subepithelial lesions (SELs) has limited diagnostic accuracy due to technical problems and small lesion size. We previously reported a novel submucosal tunneling biopsy (STB) technique for sampling SELs. This study aimed to evaluate the diagnostic ability and safety of STB compared to that of FNA for SELs. PATIENTS AND METHODS: The study was a non-randomized, prospective comparative study with crossover design in patients with endoluminal gastric SELs. Forty-three patients, including 29 cases with lesions < 2 cm were enrolled. A crossover design with 2 intervention stages (Group A: FNA followed by STB for 23 SELs, Group B: STB followed by FNA for 20 SELs) was implemented. The primary outcome was the diagnostic yield (DY). Secondary outcomes were technical success rate, procedure time, complication rate, and sample quality. RESULTS: The DY of STB was significantly higher than that of FNA (100 % vs. 34.8 %; P  < 0.0001) in group A, including 100 % in overall STB. The technical success rate of STB was significantly higher than that of FNA (100 % vs. 56.5 %; P  = 0.0006), whereas the median procedure time of STB was significantly longer than that of FNA (37 minutes vs. 18 minutes; P  < 0.0001). The median specimen area of STB samples was markedly larger than that of FNA samples (5.54 mm 2 vs. 0.69 mm 2 ; P  < 0.001). No complications occurred in either method. CONCLUSIONS: STB had significantly superior diagnostic ability and a more adequate sample quality than FNA for endoluminal gastric SELs, indicating the suitability of STB for small SELs. CLINICAL TRIAL REGISTRATION: UMIN 000006754.

12.
Int J Oncol ; 50(6): 2145-2153, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28440424

ABSTRACT

Small bowel adenocarcinoma (SBAC) accounts for 3% of all gastrointestinal tract tumors and approximately 0.5% of all cancer cases. Recent studies have indicated that the use of metformin, one of the most commonly prescribed antidiabetic drugs, is associated with a better prognosis for certain malignant diseases. However, there have been no reports on the effect of metformin in SBAC. In the present study, we evaluated the effect of metformin on human SBAC cell proliferation in vitro and in vivo and identified the microRNAs (miRNAs) associated with its antitumor effects. Metformin inhibited the proliferation of HuTu80 cells in a time- and dose-dependent manner. Importantly, metformin reduced the expression of cyclin D1, cyclin E, cyclin-dependent kinase 4, and phosphorylated retinoblastoma protein, which resulted in cell cycle arrest at the G0/G1 phase. This arrest was accompanied by activation of AMPKα and inhibition of mammalian target of rapamycin and p70s6k. Additionally, metformin reduced the levels of phosphorylated epidermal growth factor receptor and ROR2 as well as markedly altered miRNA expression in HuTu80 cells. Metformin also inhibited tumor growth in vivo in a xenograft mouse model. Our data suggest that metformin might have therapeutic potential in SBAC.


Subject(s)
Adenocarcinoma/drug therapy , Gastrointestinal Neoplasms/drug therapy , Metformin/administration & dosage , Neoplasm Proteins/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1 , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Phosphorylation , Xenograft Model Antitumor Assays
13.
Oncotarget ; 8(5): 8536-8549, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28052030

ABSTRACT

Telmisartan, a widely used antihypertensive drug, is an angiotensin II type 1 (AT1) receptor blocker (ARB). This drug inhibits cancer cell proliferation, but the underlying mechanisms in various cancers, including esophageal cancer, remain unknown. The aim of the present study was to evaluate the effects of telmisartan on human esophageal cancer cell proliferation in vitro and in vivo. We assessed the effects of telmisartan on human esophageal adenocarcinoma (EAC) cells using the cell lines OE19, OE33, and SKGT-4. Telmisartan inhibited the proliferation of these three cell lines via blockade of the G0 to G1 cell cycle transition. This blockade was accompanied by a strong decrease in cyclin D1, cyclin E, and other cell cycle-related proteins. Notably, the AMP-activated protein kinase (AMPK) pathway, a fuel sensor signaling pathway, was enhanced by telmisartan. Compound C, which inhibits the two catalytic subunits of AMPK, enhanced the expression of cyclin E, leading to G0/G1 arrest in human EAC cells. In addition, telmisartan reduced the phosphorylation of epidermal growth factor receptor (p-EGFR) and ERBB2 in vitro. In our in vivo study, intraperitoneal injection of telmisartan led to a 73.2% reduction in tumor growth in mice bearing xenografts derived from OE19 cells. Furthermore, miRNA expression was significantly altered by telmisartan in vitro and in vivo. In conclusion, telmisartan suppressed human EAC cell proliferation and tumor growth by inducing cell cycle arrest via the AMPK/mTOR pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adenocarcinoma/drug therapy , Angiotensin II Type 1 Receptor Blockers/pharmacology , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Benzoates/pharmacology , Cell Proliferation/drug effects , Esophageal Neoplasms/drug therapy , TOR Serine-Threonine Kinases/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphorylation , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Telmisartan , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
J Dig Dis ; 17(8): 518-525, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27383654

ABSTRACT

OBJECTIVES: Several studies report on the outcomes of self-expandable metallic stents (SEMSs) deployment for malignant gastric outlet obstruction (GOO). However, data was mostly based on the analysis of single-center studies including only a small number of patients. This study aimed to evaluate clinical outcomes after the deployment of SEMS in patients with malignant GOO and to compare the clinical outcomes of two metallic stents with different designs. METHODS: Altogether 125 consecutive patients from five institutions were included. Clinical outcomes were evaluated according to technical success, clinical success, stent patency period, survival period and complications. A comparison of clinical outcomes between Niti-S pyloric/duodenal and WallFlex duodenal stents was also undertaken. RESULTS: Rates for clinical and technical success were 100% and 92.0%, respectively. The median stent patency and survival periods were 72.0 days (range 3-775 days) and 75.0 days (range 3-775 days), respectively. The rate of overall adverse events was 28.8%. The rate of stent dysfunction was 16.8%, and that of adverse events, except stent dysfunction was 12.0%. Massive bleeding occurred in two patients as a late complication. The clinical success rate for Niti-S stent was significantly higher than that for WallFlex stent (96.2% vs 84.8%, P = 0.023). CONCLUSIONS: We successfully deployed a SEMS in malignant GOO. The selection of a SEMS with a lower axial force may be important for patients to resume the oral food intake. Additionally, consideration must be given to the appropriate management of fatal bleeding as a late complication.


Subject(s)
Gastric Outlet Obstruction/surgery , Neoplasms/complications , Self Expandable Metallic Stents , Adult , Aged , Aged, 80 and over , Cholangiopancreatography, Endoscopic Retrograde , Female , Gastric Outlet Obstruction/etiology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Palliative Care/methods , Prosthesis Design , Retrospective Studies , Self Expandable Metallic Stents/adverse effects , Stents , Treatment Outcome
15.
Oncol Rep ; 35(3): 1582-92, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26708419

ABSTRACT

Recent studies suggest that metformin, which is a commonly used oral anti-hyperglycemic agent of the biguanide family, may reduce cancer risk and improve prognosis, yet the detailed mechanisms by which metformin affects various types of cancers, including pancreatic cancer, remain unknown. The aim of the present study was to evaluate the effects of metformin on human pancreatic cancer cell proliferation in vitro and in vivo, and to study microRNAs (miRNAs) associated with the antitumor effect of metformin. We used the human pancreatic cancer cell lines Panc1, PK1 and PK9 to study the effects of metformin on human pancreatic cancer cells. Athymic nude mice bearing xenograft tumors were treated with or without metformin. Tumor growth was recorded after 5 weeks, and the expression of cell cycle-related proteins was determined. In addition, we used miRNA microarray tips to explore the differences in the levels of miRNAs in Panc1 cells and xenograft tumors treated with metformin or without. Metformin inhibited the proliferation of Panc1, PK1 and PK9 cells in vitro. This inhibition was accompanied by a strong decrease in G1 cyclins (particularly in cyclin D1) and retinoblastoma protein (Rb) phosphorylation. In addition, metformin reduced the phosphorylation of epidermal growth factor receptor (EGFR), particularly the phosphorylation of EGFR at Tyr845, and insulin-like growth factor 1 receptor (IGF-1R) in vitro and in vivo. miRNA expression was markedly altered by the treatment with metformin in vitro and in vivo. Our results revealed that metformin inhibits human pancreatic cancer cell proliferation and tumor growth, possibly by suppressing the cell cycle-related molecules via alteration of miRNAs.


Subject(s)
Cyclin D1/genetics , Metformin/administration & dosage , MicroRNAs/genetics , Pancreatic Neoplasms/drug therapy , Retinoblastoma Protein/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin D1/biosynthesis , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoglycemic Agents/administration & dosage , Mice , MicroRNAs/biosynthesis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphorylation , Receptor, IGF Type 1/biosynthesis , Receptor, IGF Type 1/genetics , Retinoblastoma Protein/biosynthesis , Xenograft Model Antitumor Assays
16.
Oncol Rep ; 34(6): 2987-96, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26398221

ABSTRACT

Cholangiocarcinoma (CCA) is the most common biliary malignancy and the second most common hepatic malignancy after hepatocellular carcinoma (HCC). Treatment with the anti-diabetic drug metformin has been associated with reduced cancer incidence in patients with type 2 diabetes. Thus, the present study evaluated the effects of metformin on human CCA cell proliferation in vitro and in vivo and identified the microRNAs associated with its antitumor effects. Metformin inhibited the proliferation of the CCA cell lines HuCCT-1 and TFK-1 and blocked the G0 to G1 cell cycle transition, accompanied by AMP kinase pathway activation. Metformin treatment also led to marked decreases in cyclin D1 and cyclin-dependent kinase (Cdk) 4 protein levels and retinoblastoma protein phosphorylation. However, this drug did not affect p27kip protein expression. In addition, it reduced the phosphorylation of Axl, EphA10, ALK and PYK, as well as tumor proliferation in athymic nude mice with xenograft tumors. Furthermore, it markedly altered microRNA expression. These findings suggest that metformin may have clinical use in the treatment of CCA.


Subject(s)
Cell Proliferation/drug effects , Cholangiocarcinoma/drug therapy , Metformin/administration & dosage , MicroRNAs/drug effects , Animals , Diabetes Mellitus, Type 2/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , MicroRNAs/biosynthesis , Neoplasm Proteins/biosynthesis , Xenograft Model Antitumor Assays
17.
Int J Oncol ; 47(4): 1293-302, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26252371

ABSTRACT

Although gemcitabine (2',2'-difluorocytidine monohydrochloride) is a common anticancer agent of cholangiocellular carcinoma (CCC), its growth inhibitory effects and gemcitabine resistance in CCC cells are poorly understood. Our aims were to uncover the mechanism underlying the antitumor effect of gemcitabine and to analyze the mechanism regulating in vitro CCC cell gemcitabine resistance. In addition, we sought to identify miRNAs associated with the antitumor effects of gemcitabine in CCCs. Using a cell proliferation assay and flow cytometry, we examined the ability of gemcitabine to inhibit cell proliferation in three types of human CCC cell lines (HuCCT-1, Huh28, TKKK). We also employed western blotting to investigate the effects of gemcitabine on cell cycle-related molecules in CCC cells. In addition, we used array chips to assess gemcitabine-mediated changes in angiogenic molecules and activated tyrosine kinase receptors in CCC cells. We used miRNA array chips to comprehensively analyze gemcitabine-induced miRNAs and examined clusters of differentially expressed miRNAs in cells with and without gemcitabine treatment. Gemcitabine inhibited cell proliferation in a dose- and time-dependent manner in HuCCT-1 cells, whereas cell proliferation was unchanged in Huh28 and TKKK cells. Gemcitabine inhibited cell cycle progression in HuCCT-1 cells from G0/G1 to S phase, resulting in G1 cell cycle arrest due to the reduction of cyclin D1 expression. In addition, gemcitabine upregulated the angiogenic molecules IL-6, IL-8, ENA-78 and MCP-1. In TKKK cells, by contrast, gemcitabine did not arrest the cell cycle or modify angiogenic molecules. Furthermore, in gemcitabine-sensitive HuCCT-1 cells, gemcitabine markedly altered miRNA expression. The miRNAs and angiogenic molecules altered by gemcitabine contribute to the inhibition of tumor growth in vitro.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Bile Duct Neoplasms/pathology , Cell Proliferation/drug effects , Cholangiocarcinoma/pathology , Deoxycytidine/analogs & derivatives , Blotting, Western , Cell Line, Tumor , Deoxycytidine/pharmacology , Flow Cytometry , Humans , MicroRNAs , Oligonucleotide Array Sequence Analysis , Transcriptome , Gemcitabine
18.
Oncol Rep ; 34(4): 1761-70, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26260906

ABSTRACT

Cholangiocarcinoma is the most common biliary malignancy and the second most common hepatic malignancy after hepatocellular carcinoma (HCC). Galectin-9 (Gal-9) is a tandem-repeat-type galectin that has recently been shown to exert antiproliferative effects on cancer cells. Therefore, the present study evaluated the effects of Gal-9 on the proliferation of human cholangiocarcinoma cells in vitro as well as the microRNAs (miRNAs) associated with the antitumor effects of Gal-9. Gal-9 suppressed the proliferation of cholangiocarcinoma cell lines in vitro and the growth of human cholangiocarcinoma cell xenografts in nude mice. Our data further revealed that Gal-9 increased caspase­cleaved keratin 18 (CCK18) levels, and the expression of cytochrome c increased in Gal-9-treated cholangiocarcinoma cell lines. These data suggested that Gal-9 induced cholangiocarcinoma cell apoptosis via the intrinsic apoptosis pathway mediated by caspase-dependent or -independent pathways. In addition, Gal-9 reduced the phosphorylation of the epidermal growth factor receptor (EGFR), insulin-like growth factor and insulin-like growth factor-1 receptor (IGF-1R), hepatocyte growth factor receptor and fibroblast growth factor receptor 3 (FGFR3). These findings suggest that Gal-9 can be a candidate of therapeutic target in the treatment of cholangiocarcinoma.


Subject(s)
Apoptosis/genetics , Cell Proliferation/genetics , Cholangiocarcinoma/genetics , Galectins/biosynthesis , Animals , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cholangiocarcinoma/pathology , Cytochromes c/biosynthesis , ErbB Receptors/biosynthesis , Fibroblast Growth Factors/biosynthesis , Galectins/genetics , Gene Expression Regulation, Neoplastic , Humans , Keratin-18/biosynthesis , Mice , Xenograft Model Antitumor Assays
19.
Int J Oncol ; 47(2): 535-42, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26060089

ABSTRACT

Cisplatin [cis-diamminedichloroplatinum (II)], is a platinum coordination compound that is commonly used to treat hepatocellular carcinoma (HCC). It is also one of the most compelling anticancer drugs. Recent studies suggest that cisplatin may reduce cancer risk and improve prognosis. However, the antitumor mechanism of cisplatin in several types of cancers, including HCC, has not been elucidated. The goal of the present study was to evaluate the effects of cisplatin on the proliferation of HCC cells in vitro and to determine which microRNAs (miRNAs) are associated with the anticancer effects of cisplatin in vitro. We used various human HCC-derived cell lines to study the effects of cisplatin on human HCC cells. Cisplatin led to a strong dose- and time- dependent inhibition of cell proliferation in HLE, HLF, HuH7, Li-7, Hep3B and HepG2 cells in vitro. Cisplatin also blocked the progression of the cell cycle in the G0/G1 phase, which inhibited cyclin D1 and induced apoptosis. In addition, miRNA expression was markedly altered by treatment with cisplatin in vitro. Therefore, various miRNAs induced by cisplatin may also contribute to the suppression of cellular proliferation and apoptosis. Our results demonstrate that cisplatin inhibits the growth of HCC, possibly through the induction of G1 cell cycle arrest and apoptosis through the alteration of microRNA expression.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Cisplatin/pharmacology , Liver Neoplasms/genetics , MicroRNAs/genetics , Apoptosis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , MicroRNAs/drug effects , Phosphorylation/drug effects
20.
Int J Oncol ; 46(5): 2172-80, 2015 May.
Article in English | MEDLINE | ID: mdl-25709052

ABSTRACT

Esophageal carcinoma is the eighth most common cancer worldwide and the sixth leading cause of cancer-related deaths, with one of the worst prognoses of any form of cancer. Treatment with the anti-diabetic drug metformin has been associated with reduced cancer incidence in patients with type 2 diabetes. This study therefore evaluated the effects of metformin on the proliferation, in vitro and in vivo, of human esophageal adenocarcinoma cells, as well as the microRNAs associated with the antitumor effects of metformin. Metformin inhibited the proliferation of the esophageal adenocarcinoma cell lines OE19, OE33, SK-GT4 and OACM 5.1C, blocking the G0 to G1 transition in the cell cycle. This was accompanied by strong reductions in G1 cyclins, especially cyclin D1, cyclin-dependent kinase (Cdk)4, and Cdk6, and decreases in retinoblastoma protein phosphorylation. In addition, metformin reduced the phosphorylation of epidermal growth factor receptor and insulin-like growth factor and insulin-like growth factor-1 receptor, as well as angiogenesis-related proteins, such as vascular endothelial growth factor, tissue inhibitor of metalloproteinases (TIMP)-1, and TIMP-2. Metformin also markedly altered microRNA expression. Treatment with metformin of athymic nude mice bearing xenograft tumors reduced tumor proliferation. These findings suggest that metformin may have clinical use in the treatment of esophageal adenocarcinoma.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Esophageal Neoplasms/pathology , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Disease Models, Animal , Flow Cytometry , Humans , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...