Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Carcinogenesis ; 39(9): 1127-1134, 2018 09 21.
Article in English | MEDLINE | ID: mdl-29860474

ABSTRACT

Bile duct cancer is a highly aggressive malignancy wherein early diagnosis is difficult and few treatment options are available. MicroRNA-31 (miR-31) is reported to be related with survival in patients with gastrointestinal cancers; however, the regulatory mechanism of miR-31 and association between miR-31 expression and survival in patients with bile duct cancer cases have not been established. Thus, we evaluated miR-31 expression in bile duct cancer tissues and assessed its relationship with prognosis. Additionally, we examined the effects of several cytokines on miR-31 expression. The study included 81 samples of bile duct cancer tissues. MiR-31 expression in bile duct cancer cells was significantly higher than that in normal bile duct epithelial cells (P = 0.038). There were no significant associations between miR-31 expression and clinical or pathological characteristics, except for tumour size (P = 0.012). In Kaplan-Meier analysis, high miR-31 expression was significantly associated with shorter survival (log-rank test, P = 0.0082). In multivariate Cox regression analysis, high miR-31 expression was significantly associated with prognosis (P = 0.043), independent of clinical or pathological features. Interleukin-6 (IL-6) significantly promoted miR-31 expression and cell proliferation in a dose-dependent manner, and the inhibition of STAT-3 signalling significantly suppressed miR-31 expression and cell proliferation. In conclusion, high expression was significantly associated with poor prognosis in bile duct cancer patients. The IL-6-STAT-3 signalling regulated bile duct cancer cell proliferation and miR-31 expression. Our findings suggest that miR-31 may be a promising biomarker that reflects IL-6 expression in bile duct cancer tissues and predicts poor prognosis.


Subject(s)
Bile Duct Neoplasms/genetics , Cholangiocarcinoma/genetics , Gallbladder Neoplasms/genetics , Interleukin-6/biosynthesis , MicroRNAs/genetics , Aged , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cholangiocarcinoma/mortality , Cholangiocarcinoma/pathology , Female , Gallbladder Neoplasms/mortality , Gallbladder Neoplasms/pathology , Humans , Male , Prognosis , STAT3 Transcription Factor/antagonists & inhibitors
2.
J Pathol ; 245(4): 445-455, 2018 08.
Article in English | MEDLINE | ID: mdl-29756208

ABSTRACT

RNF43 mutations are frequently detected in colorectal cancer cells and lead to a loss of function of the ubiquitin E3 ligase. Here, we investigated the clinical significance of RNF43 mutations in a large Japanese cohort and the role of RNF43 at various stages of colorectal cancer development and progression. Mutation analysis of the RNF43 gene locus with pyrosequencing technology detected RNF43 hotspot mutations in one (0.88%) of 113 colorectal polyp cases and in 30 (6.45%) of 465 colorectal cancer cases. Moreover, patients with colorectal cancer harbouring mutated RNF43 experienced a higher recurrence rate than those harbouring non-mutated RNF43. In addition, the growth of RNF43 wild-type colorectal cancer cell lines was significantly increased by RNF43 silencing. We generated Rnf43 knockout mice in a C57BL/6 N background by using the CRISPR-Cas9 system. Although intestinal organoids from Rnf43 knockout mice did not show continuous growth in the absence of R-spondin, an azoxymethane/dextran sodium sulphate mouse model demonstrated that tumours were markedly larger in Rnf43 knockout mice than in wild-type mice. These findings provide evidence that Wnt signalling activation by RNF43 mutations during the tumourigenic stage enhances tumour growth and promotes a high recurrence rate in colorectal cancer patients. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Loss of Function Mutation , Oncogene Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Aged , Animals , Biomarkers, Tumor/deficiency , Cell Movement , Cell Proliferation , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , DNA-Binding Proteins/deficiency , Disease Progression , Female , Genetic Predisposition to Disease , HCT116 Cells , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Japan , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neoplasm Recurrence, Local , Neoplasm Staging , Oncogene Proteins/deficiency , Phenotype , Risk Factors , Time Factors , Tumor Burden , Ubiquitin-Protein Ligases/deficiency , Wnt Signaling Pathway
3.
Anaerobe ; 48: 144-146, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28823592

ABSTRACT

Fusobacterium nucleatum is an invasive, adherent, and pro-inflammatory anaerobic bacterium involved in various infections and colorectal cancer. We report a case with pyogenic liver abscess, diagnosed with advanced sigmoid colon cancer, in whom F. nucleatum was simultaneously detected. In this patient, F. nucleatum was systematically analyzed using the molecular biological techniques of metagenome analysis, conventional PCR, and microbial fluorescence in situ hybridization.


Subject(s)
Fusobacterium Infections/diagnosis , Fusobacterium Infections/microbiology , Fusobacterium nucleatum/genetics , Liver Abscess, Pyogenic/diagnosis , Liver Abscess, Pyogenic/microbiology , Sigmoid Neoplasms/microbiology , Aged , Biopsy , Fusobacterium nucleatum/classification , Humans , In Situ Hybridization, Fluorescence , Male , RNA, Ribosomal, 16S , Sequence Analysis, DNA , Tomography, X-Ray Computed , Ultrasonography
4.
Carcinogenesis ; 38(4): 425-431, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28186267

ABSTRACT

Pancreatic cancer is a highly aggressive malignancy, with <50% patients surviving beyond 6 months after the diagnosis, and thus, there is an urgent need to explore new diagnostic and therapeutic approaches for this disease. Therefore, we conducted microRNA (miRNA) array analysis to detect miRNA molecules potentially associated with pancreatic cancer malignancy. To assess the identified miRNAs, we performed quantitative reverse transcription-PCR on 248 pancreatic ductal adenocarcinomas (UICC stage II). We also examined miRNA expression [microRNA-21 (miR-21) and microRNA-31 (miR-31)] and epigenetic alterations, including CpG island methylator phenotype (CIMP), potentially associated with the identified miRNAs. For functional analysis, we conducted proliferation and invasion assays using a pancreatic cancer cell line. miRNA array analysis revealed that microRNA-196b (miR-196b) was the most up-regulated miRNA in pancreatic cancer tissues compared with normal pancreatic duct cells. High miR-196b expression was associated with miR-21 (P = 0.0025) and miR-31 (P = 0.0001) expression. It was also related to poor prognosis in the multivariate analysis using overall survival (hazard ratio: 1.66; 95% confidence interval: 1.09-2.54; P = 0.019). Functional analysis demonstrated that miR-196b inhibitor decreased cell proliferation and that miR-196b mimic promoted cancer cell invasion. In conclusion, a significant association of high miR-196b expression with poor prognosis was observed in pancreatic cancer. Our data also revealed that miR-196b played an oncogenic role and that the transfection of the miR-196b inhibitor had an anti-tumour effect in the pancreatic cancer cell line. These results suggest that miR-196b is a promising diagnostic biomarker and therapeutic target in pancreatic cancer.


Subject(s)
Biomarkers, Tumor/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Aged , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Humans , Kaplan-Meier Estimate , Male , Neoplasm Invasiveness/genetics , Prognosis , Proportional Hazards Models , Up-Regulation/genetics
5.
Oncotarget ; 8(11): 17810-17818, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28147317

ABSTRACT

The polycomb group protein enhancer of zeste homolog 2 (EZH2) is a methyltransferase that suppresses microRNA-31 (miR-31) in various human malignancies including colorectal cancer. We recently suggested that miR-31 regulates the signaling pathway downstream of epidermal growth factor receptor (EGFR) in colorectal cancer. Therefore, we conducted this study for assessing the relationship between EZH2 expression and clinical outcomes in patients with colorectal cancer treated with anti-EGFR therapeutics. We immunohistochemically evaluated EZH2 expression and assessed miR-31 and gene mutations [KRAS (codon 61/146), NRAS (codon 12/13/61), and BRAF (codon 600)] in 109 patients with colorectal cancer harboring KRAS (codon 12/13) wild-type. We also evaluated the progression-free survival (PFS) and overall survival (OS). In the result, low EZH2 expression was significantly associated with shorter PFS (log-rank test: P = 0.023) and OS (P = 0.036) in patients with colorectal cancer. In the low-miR-31-expression group and the KRAS (codon 61/146), NRAS, and BRAF wild-type groups, a significantly shorter PFS (P = 0.022, P = 0.039, P = 0.021, and P = 0.036, respectively) was observed in the EZH2 low-expression groups than in the high-expression groups. In the multivariate analysis, low EZH2 expression was associated with a shorter PFS (P = 0.046), independent of the mutational status and miR-31. In conclusion, EZH2 expression was associated with survival in patients with colorectal cancer who were treated with anti-EGFR therapeutics. Moreover, low EZH2 expression was independently associated with shorter PFS in patients with cancer, suggesting that EZH2 expression is a useful additional prognostic biomarker for anti-EGFR therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Enhancer of Zeste Homolog 2 Protein/metabolism , ErbB Receptors/antagonists & inhibitors , Antibodies, Monoclonal/therapeutic use , Biomarkers, Tumor/biosynthesis , Cetuximab/therapeutic use , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Disease-Free Survival , Enhancer of Zeste Homolog 2 Protein/biosynthesis , Female , GTP Phosphohydrolases/genetics , Humans , Male , Membrane Proteins/genetics , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , Panitumumab , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Treatment Outcome
6.
Oncotarget ; 7(11): 12704-17, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26871294

ABSTRACT

Polycomb group protein enhancer of zeste homolog 2 (EZH2) is a methyltransferase that correlates with the regulation of invasion and metastasis and is overexpressed in human cancers such as colorectal cancer. MicroRNA-31 (miR-31) plays an oncogenic role and is associated with BRAF mutation and poor prognosis in colorectal cancer. EZH2 is functionally considered to suppress miR-31 expression in human cancers; however, no study has reported its relationship with colon cancer. We therefore evaluated EZH2 expression using immunohistochemistry and assessed miR-31 and epigenetic alterations using 301 colorectal carcinomas and 207 premalignant lesions. Functional analysis was performed to identify the association between EZH2 and miR-31 using cancer cell lines. In the current study, negative, weak, moderate, and strong EZH2 expressions were observed in 15%, 19%, 25%, and 41% of colorectal cancers, respectively. EZH2 was inversely associated with miR-31 (P < 0.0001), independent of clinicopathological and molecular features. In a multivariate stage-stratified analysis, high EZH2 expression was related to favorable prognosis (P = 0.0022). Regarding premalignant lesions, negative EZH2 expression was frequently detected in sessile serrated adenomas/polyps (SSA/Ps) (76%; P < 0.0001) compared with hyperplastic polyps, traditional serrated adenomas, and non-serrated adenomas (25-36%). Functional analysis demonstrated that the knockdown of EZH2 increased miR-31 expression. In conclusion, an inverse association was identified between EZH2 and miR-31 in colorectal cancers. Our data also showed that upregulation of EZH2 expression may be rare in SSA/Ps. These results suggest that EZH2 suppresses miR-31 in colorectal cancer and may correlate with differentiation and evolution of serrated pathway.


Subject(s)
Colorectal Neoplasms/pathology , Enhancer of Zeste Homolog 2 Protein/metabolism , MicroRNAs/metabolism , Precancerous Conditions/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenomatous Polyps/genetics , Adenomatous Polyps/metabolism , Adenomatous Polyps/pathology , Adult , Aged , Colonic Polyps/genetics , Colonic Polyps/metabolism , Colonic Polyps/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Kaplan-Meier Estimate , Male , MicroRNAs/genetics , Middle Aged , Precancerous Conditions/genetics , Precancerous Conditions/metabolism
7.
World J Gastroenterol ; 22(2): 557-66, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26811607

ABSTRACT

The human intestinal microbiome plays a major role in human health and diseases, including colorectal cancer. Colorectal carcinogenesis represents a heterogeneous process with a differing set of somatic molecular alterations, influenced by diet, environmental and microbial exposures, and host immunity. Fusobacterium species are part of the human oral and intestinal microbiota. Metagenomic analyses have shown an enrichment of Fusobacterium nucleatum (F. nucleatum) in colorectal carcinoma tissue. Using 511 colorectal carcinomas from Japanese patients, we assessed the presence of F. nucleatum. Our results showed that the frequency of F. nucleatum positivity in the Japanese colorectal cancer was 8.6% (44/511), which was lower than that in United States cohort studies (13%). Similar to the United States studies, F. nucleatum positivity in Japanese colorectal cancers was significantly associated with microsatellite instability (MSI)-high status. Regarding the immune response in colorectal cancer, high levels of infiltrating T-cell subsets (i.e., CD3+, CD8+, CD45RO+, and FOXP3+ cells) have been associated with better patient prognosis. There is also evidence to indicate that molecular features of colorectal cancer, especially MSI, influence T-cell-mediated adaptive immunity. Concerning the association between the gut microbiome and immunity, F. nucleatum has been shown to expand myeloid-derived immune cells, which inhibit T-cell proliferation and induce T-cell apoptosis in colorectal cancer. This finding indicates that F. nucleatum possesses immunosuppressive activities by inhibiting human T-cell responses. Certain microRNAs are induced during the macrophage inflammatory response and have the ability to regulate host-cell responses to pathogens. MicroRNA-21 increases the levels of IL-10 and prostaglandin E2, which suppress antitumor T-cell-mediated adaptive immunity through the inhibition of the antigen-presenting capacities of dendritic cells and T-cell proliferation in colorectal cancer cells. Thus, emerging evidence may provide insights for strategies to target microbiota, immune cells and tumor molecular alterations for colorectal cancer prevention and treatment. Further investigation is needed to clarify the association of Fusobacterium with T-cells and microRNA expressions in colorectal cancer.


Subject(s)
Adaptive Immunity , Biomarkers, Tumor/genetics , Colorectal Neoplasms/microbiology , Fusobacterium Infections/microbiology , Fusobacterium nucleatum/pathogenicity , Gastrointestinal Microbiome , Immunity, Innate , Animals , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Fusobacterium Infections/genetics , Fusobacterium Infections/immunology , Fusobacterium Infections/metabolism , Fusobacterium nucleatum/immunology , Fusobacterium nucleatum/metabolism , Host-Pathogen Interactions , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/microbiology , Microsatellite Instability , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , Tumor Escape , Tumor Microenvironment
8.
Oncotarget ; 6(26): 22114-25, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26090613

ABSTRACT

Although gastrointestinal carcinoid tumors are relatively rare in the digestive tract, a quarter of them are present in the rectum. In the absence of specific tumor biomarkers, lymphatic or vascular invasion is generally used to predict the risk of lymph node metastasis. We, therefore, examined the genetic and epigenetic alterations potentially associated with lymphovascular invasion among 56 patients with rectal carcinoid tumors. We also conducted a microRNA (miRNA) array analysis. Our analysis failed to detect mutations in BRAF, KRAS, NRAS, or PIK3CA or any microsatellite instability (MSI); however, we did observe CpG island methylator phenotype (CIMP) positivity in 13% (7/56) of the carcinoid tumors. The CIMP-positive status was significantly correlated with lymphovascular invasion (P = 0.036). The array analysis revealed that microRNA-885 (miR-885)-5p was the most up-regulated miRNA in the carcinoid tumors with lymphovascular invasion compared with that in those without invasion. In addition, high miR-885-5p expression was independently associated with lymphovascular invasion (P = 0.0002). In conclusion, our findings suggest that miR-885-5p and CIMP status may be useful biomarkers for predicting biological malignancy in patients with rectal carcinoid tumors.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoid Tumor/genetics , Rectal Neoplasms/genetics , Biomarkers, Tumor/biosynthesis , Carcinoid Tumor/pathology , Female , Humans , Lymphatic Metastasis , Male , MicroRNAs/biosynthesis , MicroRNAs/genetics , Middle Aged , Prognosis , Rectal Neoplasms/pathology
9.
Oncotarget ; 6(9): 7209-20, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25797243

ABSTRACT

Recently, bacterial infection causing periodontal disease has attracted considerable attention as a risk factor for pancreatic cancer. Fusobacterium species is an oral bacterial group of the human microbiome. Some evidence suggests that Fusobacterium species promote colorectal cancer development; however, no previous studies have reported the association between Fusobacterium species and pancreatic cancer. Therefore, we examined whether Fusobacterium species exist in pancreatic cancer tissue. Using a database of 283 patients with pancreatic ductal adenocarcinoma (PDAC), we tested cancer tissue specimens for Fusobacterium species. We also tested the specimens for KRAS, NRAS, BRAF and PIK3CA mutations and measured microRNA-21 and microRNA-31. In addition, we assessed epigenetic alterations, including CpG island methylator phenotype (CIMP). Our data showed an 8.8% detection rate of Fusobacterium species in pancreatic cancers; however, tumor Fusobacterium status was not associated with any clinical and molecular features. In contrast, in multivariate Cox regression analysis, compared with the Fusobacterium species-negative group, we observed significantly higher cancer-specific mortality rates in the positive group (p = 0.023). In conclusion, Fusobacterium species were detected in pancreatic cancer tissue. Tumor Fusobacterium species status is independently associated with a worse prognosis of pancreatic cancer, suggesting that Fusobacterium species may be a prognostic biomarker of pancreatic cancer.


Subject(s)
Fusobacterium Infections/complications , Fusobacterium/pathogenicity , Pancreatic Neoplasms/microbiology , Aged , Biomarkers, Tumor/metabolism , CpG Islands , DNA Methylation , Epigenesis, Genetic , Female , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Male , Microsatellite Instability , Middle Aged , Multivariate Analysis , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/genetics , Phenotype , Prognosis , Proportional Hazards Models , Regression Analysis , Sequence Analysis, DNA , Treatment Outcome
10.
Int J Cancer ; 137(6): 1258-68, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-25703934

ABSTRACT

Human gut microbiota is being increasingly recognized as a player in colorectal cancers (CRCs). Evidence suggests that Fusobacterium nucleatum (F. nucleatum) may contribute to disease progression and is associated with CpG island methylator phenotype (CIMP) and microsatellite instability (MSI) in CRCs; however, to date, there are no reports about the relationship between F. nucleatum and molecular features in the early stage of colorectal tumorigenesis. Therefore, we investigated the presence of F. nucleatum in premalignant colorectal lesions. In total, 465 premalignant lesions (343 serrated lesions and 122 non-serrated adenomas) and 511 CRCs were studied. We determined the presence of F. nucleatum and analyzed its association with molecular features including CIMP, MSI and microRNA-31 status. F. nucleatum was detected in 24% of hyperplastic polyps, 35% of sessile serrated adenomas (SSAs), 30% of traditional serrated adenomas (TSAs) and 33% of non-serrated adenomas. F. nucleatum was more frequently detected in CIMP-high premalignant lesions than in CIMP-low/zero lesions (p = 0.0023). In SSAs, F. nucleatum positivity increased gradually from sigmoid colon to cecum (p = 0.042). F. nucleatum positivity was significantly higher in CRCs (56%) than in premalignant lesions of any histological type (p < 0.0001). In conclusion, F. nucleatum was identified in premalignant colorectal lesions regardless of histopathology but was more frequently associated with CIMP-high lesions. Moreover, F. nucleatum positivity increased according to histological grade, suggesting that it may contribute to the progression of colorectal neoplasia. Our data also indicate that F. nucleatum positivity in SSAs may support the "colorectal continuum" concept.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/microbiology , Fusobacterium Infections/genetics , Fusobacterium Infections/pathology , Fusobacterium nucleatum/isolation & purification , Aged , Cecum/microbiology , Cecum/pathology , Colon/microbiology , Colon/pathology , Colorectal Neoplasms/pathology , CpG Islands/genetics , DNA Methylation/genetics , Female , Fusobacterium Infections/microbiology , Humans , Male , MicroRNAs/genetics , Microsatellite Instability
11.
Digestion ; 91(1): 57-63, 2015.
Article in English | MEDLINE | ID: mdl-25632919

ABSTRACT

BACKGROUND: The population in Japan is aging more rapidly than in any other country. However, no studies have determined the characteristics of the large population of elderly patients with colorectal tumors. Therefore, we examined the clinicopathological and molecular features of these tumors in elderly patients. METHODS: In total, 1,627 colorectal tumors (393 serrated lesions, 277 non-serrated adenomas and 957 colorectal cancers) were acquired from patients. Tumor specimens were analyzed for BRAF and KRAS mutations, CpG island methylator phenotype-specific promoters (CACNA1G, CDKN2A, IGF2 and RUNX3), IGFBP7, MGMT, MLH1 and RASSF2 methylation, microsatellite instability (MSI) and microRNA- 31 (miR-31). RESULTS: The frequency of elderly patients (aged ≥75 years) with sessile serrated adenomas (SSAs) with cytological dysplasia was higher than that of those with other serrated lesions and non-serrated adenomas (p < 0.0001). In elderly patients, all SSAs were located in the proximal colon (particularly the cecum to ascending colon). High miR-31 expression, MLH1 methylation and MSI-high status were more frequently detected in SSAs from elderly patients than in those from non-elderly patients. In contrast, no significant differences were found between older age of onset and high-grade dysplasia for traditional serrated adenomas or non-serrated adenomas in any of these molecular alterations. CONCLUSION: In elderly patients, all SSAs were located in the proximal colon. Furthermore, cytological dysplasia and molecular alterations were more frequently detected in elderly patients with SSAs than in non-elderly patients. Thus, careful colonoscopic examinations of the proximal colon are necessary for elderly patients because SSAs in those patients may exhibit malignant potential.


Subject(s)
Adenoma , Colon/pathology , Colonic Polyps , Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Adenoma/genetics , Adenoma/pathology , Aged , Aged, 80 and over , Asian People , Colonic Polyps/classification , Colonic Polyps/genetics , Colonic Polyps/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation , Female , Humans , Hyperplasia , Japan , Male , Microsatellite Instability , Mutation , Phenotype
12.
Ann Surg Oncol ; 22(8): 2640-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25472647

ABSTRACT

BACKGROUND: Gene mutations in the pathway downstream of epidermal growth factor receptor (EGFR) are considered to induce resistance to anti-EGFR therapy in colorectal cancer (CRC). We recently reported that microRNA-31 (miR-31)-5p may regulate BRAF activation and play a role in the signaling pathway downstream of EGFR in CRC. Therefore, we hypothesized that miR-31-5p can be a useful biomarker for anti-EGFR therapy in CRC. METHODS: We evaluated miR-31-5p expression and gene mutations [KRAS (codon 61 or 146), NRAS (codon 12, 13, or 61), and BRAF (V600E)] in the EGFR downstream pathway in 102 CRC patients harboring KRAS (codon 12 or 13) wild-type who were treated with anti-EGFR therapeutics. Progression-free survival (PFS) and overall survival (OS) were evaluated. RESULTS: KRAS (codon 61 or 146), NRAS, and BRAF mutations were detected in 6.9, 6.9, and 5.9 % patients, respectively. Compared with CRCs with at least one mutation (n = 20), significantly better PFS (P = 0.0003) but insignificantly better OS were observed in CRCs harboring all wild-type genes (KRAS, NRAS, and BRAF). High miR-31-5p expression was identified in 11 % (n = 11) patients and was significantly associated with shorter PFS (P = 0.003). In CRCs carrying all wild-type genes, high miR-31-5p was associated with shorter PFS (P = 0.027). CONCLUSIONS: High miR-31-5p expression was associated with shorter PFS in patients with CRC treated with anti-EGFR therapeutics. Moreover, in CRCs carrying all wild-type genes, high miR-31-5p was associated with shorter PFS, suggesting that it may be a useful and additional prognostic biomarker for anti-EGFR therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , ErbB Receptors/antagonists & inhibitors , MicroRNAs/genetics , Aged , Antibodies, Monoclonal/therapeutic use , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cetuximab/therapeutic use , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Disease-Free Survival , Female , GTP Phosphohydrolases/genetics , Gene Expression , Humans , Male , Membrane Proteins/genetics , Middle Aged , Neoplasm Metastasis , Panitumumab , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Response Evaluation Criteria in Solid Tumors , Survival Rate
13.
Intern Med ; 53(18): 2121-5, 2014.
Article in English | MEDLINE | ID: mdl-25224200

ABSTRACT

Plasma cell leukemia (PCL) is a rare variant of multiple myeloma (MM) with a poor prognosis. Nonsecretory myeloma is also a rare form of MM characterized by the absence of detectable M-protein in the serum and urine. This report describes two cases of nonsecretory PCL. The first patient was an 85-year-old man in whom the lack of monoclonal immunoglobulins made it difficult to make a diagnosis because the malignant cells showed an atypical morphology. He died of rapid disease progression before starting chemotherapy. The second patient was a 78-year-old woman whose tumor cells displayed a typical plasma cell morphology. She was successfully treated with bortezomib-containing chemotherapy.


Subject(s)
Leukemia, Plasma Cell/diagnosis , Multiple Myeloma/diagnosis , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Biopsy , Diagnosis, Differential , Disease Progression , Fatal Outcome , Female , Humans , Leukemia, Plasma Cell/drug therapy , Male , Multiple Myeloma/drug therapy
14.
Gan To Kagaku Ryoho ; 39(10): 1563-6, 2012 Oct.
Article in Japanese | MEDLINE | ID: mdl-23064073

ABSTRACT

S-1 and capecitabine are orally administered fluoropyridines reported to be effective in the treatment of advanced gastric cancer(AGC). In fact, both S-1/CDDP and capecitabine/CDDP are considered to be the standard first-line treatments for AGC.However, no information concerning on the activity of capecitabine in S-1-pretreated patients with AGC has been reported. Here, we present a case of recurrent gastric cancer that showed a partial response resulting in 6 months of progres-sion-free survival, thanks to capecitabine/CDDP after the failure of multiple anticancer drugs such as S-1/CDDP. S -1 and capecitabine may exhibit cross-resistance because they both have the same final active metabolite: 5-fluorouracil(5-FU). Dihydropyrimidine dehydrogenase(DPD)is the rate-limiting enzyme in the degradation of 5-FU, and S-1 contains the inhibitor of DPD. Thus, S-1, but not capecitabine, is active against tumors with high DPD expression. On the other hand, capecitabine is activated to 5-FU by thymidine phosphorylase(TP)within the tumor tissue and is more effective against tumors with high TP expression. The present case suggests that S-1 and capecitabine do not always exhibit cross-resistance, and that capecitabine may be effective in S-1-pretreated patients with AGC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Stomach Neoplasms/drug therapy , Aged , Capecitabine , Cisplatin/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Combinations , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Humans , Male , Oxonic Acid/administration & dosage , Recurrence , Salvage Therapy , Tegafur/administration & dosage
15.
Gan To Kagaku Ryoho ; 37(5): 891-4, 2010 May.
Article in Japanese | MEDLINE | ID: mdl-20495322

ABSTRACT

A 75-year-old male who with type 3 gastric cardia cancer with multiple liver metastases was initially treated with S-1 in July of 2005. After 4 courses of the treatment, the liver metastases became undetectable on abdominal CT scan, with reduction in size of the primary tumor of the stomach. After 7 months of S-1 treatment, however, the progression of the primary lesion was endoscopically detected, and irinotecan was administered, demonstrating primary tumor regression. When re-growth of the primary tumor was observed, 3 courses of paclitaxel treatment showed little effect and was replaced by docetaxel treatment for 5 months, which had a grade 3 adverse effect. The next 10 courses of 5-FU combined with methotrexate were applied for one year until the primary tumor showed enlargement. Then 12 courses of CDDP with S-1 were administered until now, and the size of the primary carcinoma is under control. The patient is being followed on an outpatient basis without any surgical treatment, while the liver metastases have not relapsed on abdominal imaging.


Subject(s)
Antineoplastic Agents/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Aged , Gastroscopy , Humans , Liver Neoplasms/diagnostic imaging , Male , Neoplasm Staging , Quality of Life , Stomach Neoplasms/diagnostic imaging , Time Factors , Tomography, X-Ray Computed
16.
Biochem Biophys Res Commun ; 367(3): 674-9, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18174021

ABSTRACT

Galantamine, a reversible inhibitor of acetylcholine esterase (AChE), is a novel drug treatment for mild to moderate Alzheimer's disease and vascular dementia. Interestingly, it has been suggested that galantamine treatment is associated with more clinical benefit in patients with mild-to-moderate Alzheimer disease compared to other AChE inhibitors. We hypothesized that the protective effects of galantamine would involve induction of the protective gene, heme oxygenase-1 (HO-1), in addition to enhancement of the cholinergic system. Brain microvascular endothelial cells (mvECs) were isolated from spontaneous hypertensive rats. Galantamine significantly reduced H(2)O(2)-induced cell death of mvECs in association with HO-1 induction. These protective effects were completely reversed by nuclear factor-kappaB (NF-kappaB) inhibition or HO inhibition. Furthermore, galantamine failed to induce HO-1 in mvECs which lack inducible nitric oxide synthase (iNOS), supplementation of a nitric oxide (NO) donor or iNOS gene transfection on iNOS-deficient mvECs resulted in HO-1 induction with galantamine. These data suggest that the protective effects of galantamine require NF-kappaB activation and iNOS expression, in addition to HO-1. Likewise, carbon monoxide (CO), one of the byproducts of HO, up-regulated HO-1 and protected mvECs from oxidative stress in a similar manner. Our data demonstrate that galantamine mediates cytoprotective effects on mvECs through induction HO-1. This pharmacological action of galantamine may, at least in part, account for the superior clinical efficacy of galantamine in vascular dementia and Alzheimer disease.


Subject(s)
Brain/blood supply , Carbon Monoxide/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Galantamine/pharmacology , Heme Oxygenase-1/metabolism , Animals , Cell Death/drug effects , Cells, Cultured , Cytoprotection/drug effects , Electrophoretic Mobility Shift Assay , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Gene Transfer Techniques , Heme Oxygenase-1/antagonists & inhibitors , Humans , Hydrogen Peroxide/toxicity , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Oxidants/toxicity , Oxidative Stress/drug effects , Rats , Rats, Inbred SHR , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
17.
Clin Exp Pharmacol Physiol ; 33(7): 623-7, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16789930

ABSTRACT

1. Low-voltage electrical stimulation (LVES) in skeletal muscle at a level far below the threshold of muscle contraction has been reported to promote local angiogenesis. However, the mechanism underlying the promotion of local angiogenesis by LVES has not been fully elucidated. In the present study, we evaluated whether angiogenic factors, such as vascular endotherial growth factor (VEGF), hepatocyte growth factor (HGF) and fibroblast growth factor (FGF), as well as other disadvantageous factors, such as inflammation (interleukin (IL)-6) and hypoxia (hypoxia-inducible factor (HIF)-1alpha), contribute to the local angiogenesis produced by LVES. 2. We completely excised bilateral femoral arteries of male Sprague-Dawley rats. After the operation, electrodes were implanted onto the centre of the fascia of the bilateral tibialis anterior (TA) muscles, tunnelled subcutaneously and exteriorized at the level of the scapulae. The right TA muscles of rats were stimulated continuously at a stimulus frequency of 50 Hz, with a 0.1 V stimulus strength and no interval, for 5 days. The left TA muscles served as controls. 3. We found that both VEGF and HGF protein were significantly increased by LVES in stimulated muscles compared with control. The VEGF level of the LVES group was 89.10 +/- 17.19 ng/g, whereas that of the control group was 65.07 +/- 12.88 ng/g, as determined by ELISA (P < 0.05). The HGF level of the LVES and control groups was 8.52 +/- 1.96 and 5.80 +/- 2.14 ng/g, respectively (P < 0.05). In contrast, there was no difference in FGF, IL-6 and HIF-1alpha between the LVES and control groups. 4. These results suggest that LVES in a hindlimb ischaemia model of rats increases not only VEGF, but also HGF, production, which may be the main mechanism responsible for the angiogenesis produced by LVES.


Subject(s)
Angiogenic Proteins/metabolism , Hepatocyte Growth Factor/metabolism , Ischemia/metabolism , Muscle, Skeletal/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Electric Stimulation , Enzyme-Linked Immunosorbent Assay , Hindlimb , Immunohistochemistry , Male , Muscle, Skeletal/blood supply , Rats , Rats, Sprague-Dawley , Up-Regulation
18.
Circulation ; 112(4): 587-91, 2005 Jul 26.
Article in English | MEDLINE | ID: mdl-16027253

ABSTRACT

BACKGROUND: Autologous vein grafts and balloon angioplasty are still commonly used for arterial reconstructive procedures. Their success is limited by the development of intimal hyperplasia (IH). Biliverdin (BVD), one of the by-products of heme degradation, has been shown to have potent antioxidant and antiinflammatory effects. We hypothesized that BVD administration would protect vascular tissue against vascular injury. METHODS AND RESULTS: The effects of BVD administration against IH after vascular injury were analyzed in an arterialized vein graft model and a balloon injury model in rats. BVD treatment significantly suppressed the development of IH in both models compared with those without BVD. The mechanisms by which BVD treatment inhibits IH development might include decreasing c-Jun NH2 terminal kinase activation and preventing apoptosis of endothelial cells. BVD also suppressed vascular smooth muscle cell migration in vitro. CONCLUSIONS: BVD administration prevented IH associated with arterialized vein graft vasculopathy or balloon angioplasty-induced vessel injury. These results suggest that a treatment regimen with exogenous BVD administration could provide an effective therapeutic adjunct to facilitate transfer of experimental treatments for vascular injury to the clinic.


Subject(s)
Biliverdine/pharmacology , Tunica Intima/pathology , Vascular Diseases/drug therapy , Animals , Apoptosis , Cell Movement/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Hyperplasia , JNK Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Phosphorylation , Rats , Rats, Inbred Lew , Vascular Diseases/pathology
19.
Am J Transplant ; 5(2): 282-91, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15643987

ABSTRACT

Both carbon monoxide (CO) and biliverdin, products of heme degradation by heme oxygenase, have been shown to attenuate ischemia/reperfusion (I/R) injury. We hypothesized in this study that dual-treatment with CO and biliverdin would induce enhanced protective effects against cold I/R injury. Heterotopic heart and orthotopic kidney transplantation were performed in syngeneic Lewis rats after 24-h cold preservation in UW solution. While monotherapy with CO (20 ppm) or biliverdin (50 mg/kg, ip) did not alter the survival of heart grafts, dual-treatment increased survival to 80% from 0% in untreated recipients, with a significant decrease of myocardial injury and improved cardiac function. Similarly, dual-treatment significantly improved glomerular filtration rates of renal grafts and prolonged recipient survival compared to untreated controls. I/R injury-induced up-regulation of pro-inflammatory mediators (e.g. TNF-alpha, iNOS) and extravasation of inflammatory infiltrates were significantly less with dual-treatment than untreated controls. In addition, dual-treatment was effective in decreasing lipid peroxidation and improving graft blood flow through the distinctive action of biliverdin and CO, respectively. The study shows that the addition of byproducts of heme degradation with different mechanisms of action provides enhanced protection against transplant-associated cold I/R injury of heart and kidney grafts.


Subject(s)
Biliverdine/therapeutic use , Carbon Monoxide/therapeutic use , Heart Transplantation , Kidney Transplantation , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Animals , Cell Movement/physiology , Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1 , Macrophages/physiology , Male , Oxygen/metabolism , Rats , Rats, Inbred Lew
20.
Proc Natl Acad Sci U S A ; 101(33): 12277-81, 2004 Aug 17.
Article in English | MEDLINE | ID: mdl-15304656

ABSTRACT

Stem cell therapy holds great promise for the replacement of damaged or dysfunctional myocardium. Nitric oxide (NO) has been shown to promote embryonic stem (ES) cell differentiation in other systems. We hypothesized that NO, through NO synthase gene transfer or exogenous NO exposure, would promote the differentiation of mouse ES cells into cardiomyocytes (CM). In our study, NO treatment increased both the number and the size of beating foci in embryoid body (EB) outgrowths. Within 2 weeks, 69% of the inducible NO synthase-transduced EB displayed spontaneously beating foci, as did 45% of the NO donor-treated EB, compared with only approximately 15% in controls. Cardiac-specific genes and protein expression were significantly increased in NO-treated ES. Electron microscopy and immunocytochemistry revealed that these NO-induced contracting cells exhibited characteristics consistent with CM. At day 7 in culture, troponin T was expressed in 45.6 +/- 20.6% of the NO-treated ES cells but in only 9.25 +/- 1.77% of control cells. Interestingly, 50.4 +/- 18.4% of NO-treated ES cells were troponin T-negative and annexin V-positive. This apoptotic phenotype was seen in <1% of the control ES cells. These data strongly support our hypothesis that mouse ES cells can be accelerated to differentiate into CM by NO treatment. NO may influence cardiac differentiation by both inducing a switch toward a cardiac phenotype and inducing apoptosis in cells not committed to cardiac differentiation.


Subject(s)
Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Nitric Oxide/pharmacology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line , Gene Expression Regulation, Developmental/drug effects , Mice , Microscopy, Electron , Myocytes, Cardiac/metabolism , Phenotype , Pluripotent Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...