Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
2.
Tohoku J Exp Med ; 238(2): 131-41, 2016 02.
Article in English | MEDLINE | ID: mdl-26829886

ABSTRACT

Glucose is a major energy source for mammalian cells and is transported into cells via cell-specific expression of various glucose transporters (GLUTs). Especially, cancer cells require massive amounts of glucose as an energy source for their dysregulated growth and thus over-express GLUTs. d-allose, a C-3 epimer of d-glucose, is one of rare sugars that exist in small quantities in nature. We have shown that d-allose induces the tumor suppressor gene coding for thioredoxin interacting protein (TXNIP) and inhibits cancer cell growth by G1 cell cycle arrest. It has also been reported that GLUTs including GLUT1 are over-expressed in many cancer cell lines, which may contribute to larger glucose utilization. Since d-allose suppresses the growth of cancer cells through the upregulation of TXNIP expression, our present study focused on whether d-allose down-regulates GLUT1 expression via TXNIP expression and thus suppresses cancer cell growth. Western blot and real-time PCR analyses revealed that d-allose significantly induced TXNIP expression and inhibited GLUT1 expression in a dose-dependent manner in three human cancer cell lines: hepatocellular carcinoma (HuH-7), Caucasian breast adenocarcinoma (MDA-MB-231), and neuroblastoma (SH-SY5Y). In these cell lines, d-allose treatment inhibited cell growth. Importantly, d-allose treatment decreased glucose uptake, as measured by the uptake of 2-deoxy d-glucose. Moreover, the reporter assays showed that d-allose decreased the expression of luciferase through the hypoxia response element present in the tested promoter region. These results suggest that d-allose may cause the inhibition of cancer growth by reducing both GLUT1 expression and glucose uptake.


Subject(s)
Glucose Transporter Type 1/genetics , Glucose/pharmacology , Neoplasms/genetics , Neoplasms/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements , Up-Regulation/drug effects
3.
J Neuroinflammation ; 11: 173, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25311735

ABSTRACT

BACKGROUND: The cochlea is the sensory organ of hearing. In the cochlea, the organ of Corti houses sensory cells that are susceptible to pathological insults. While the organ of Corti lacks immune cells, it does have the capacity for immune activity. We hypothesized that resident cells in the organ of Corti were responsible for the stress-induced immune response of the organ of Corti. This study profiled the molecular composition of the immune system in the organ of Corti and examined the immune response of non-immune epithelial cells to acoustic overstimulation. METHODS: Using high-throughput RNA-sequencing and qRT-PCR arrays, we identified immune- and inflammation-related genes in both the cochlear sensory epithelium and the organ of Corti. Using bioinformatics analyses, we cataloged the immune genes expressed. We then examined the response of these genes to acoustic overstimulation and determined how changes in immune gene expression were related to sensory cell damage. RESULTS: The RNA-sequencing analysis reveals robust expression of immune-related genes in the cochlear sensory epithelium. The qRT-PCR array analysis confirms that many of these genes are constitutively expressed in the resident cells of the organ of Corti. Bioinformatics analyses reveal that the genes expressed are linked to the Toll-like receptor signaling pathway. We demonstrate that expression of Toll-like receptor signaling genes is predominantly from the supporting cells in the organ of Corti cells. Importantly, our data demonstrate that these Toll-like receptor pathway genes are able to respond to acoustic trauma and that their expression changes are associated with sensory cell damage. CONCLUSION: The cochlear resident cells in the organ of Corti have immune capacity and participate in the cochlear immune response to acoustic overstimulation.


Subject(s)
Cytokines/metabolism , Gene Expression Regulation/physiology , Hearing Disorders/pathology , Organ of Corti/pathology , Sensory Receptor Cells/metabolism , Signal Transduction/genetics , Toll-Like Receptors/metabolism , Acoustic Stimulation , Animals , Computational Biology , Cytokines/genetics , Epithelial Cells/metabolism , Evoked Potentials, Auditory, Brain Stem/physiology , Genotype , Hearing Disorders/etiology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Noise/adverse effects , Toll-Like Receptor 4/genetics , Toll-Like Receptors/genetics
4.
J Transl Med ; 12: 150, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24884939

ABSTRACT

BACKGROUND: Previous reports showed the presence of limited numbers of stem cells in neonatal murine cochlear sensory epithelia and these cells are progressively lost during the postnatal development. The goal of this study was to investigate whether stem cells can be derived from mature mouse cochleae under suspension culture conditions, and to analyze the expression of the stem cell and inner ear progenitor cell markers in cells dissociated from neonatal and adult mouse organs of Corti. METHODS: Organs of Corti were dissected from postnatal day 1 (P1) or postnatal day 60 (P60) mouse. The dissociated cells were cultivated under suspension cultures conditions. Reverse transcription-polymerase chain reaction (RT-PCR) and immunocytochemistry were conducted for phenotype characterization. RESULTS: The number of cochlear stem cells (otospheres) yielded from P1 organ of Corti was significantly higher than that of the P60 organ of Corti. RT-PCR analyses showed that the stem markers, such as nanog, sox2, klf4, and nestin can be found to be distributed similarly in the cells derived from both of organisms, but the inner ear developmental/progenitor cell markers showed lower expression in P60 organ of Corti compared to P1. Immunocytochemistry results also revealed the evidence that P60 otospheres lacking of differentiation potential in vitro, which opposed to the strong differentiation potential of otospheres at P1 stage. CONCLUSIONS: Our findings suggest that the loss of numbers and features of stem cells in the adult organ of Corti is associated with the substantial down-regulation of inner ear progenitor key-markers during maturation of the cells in organ of Corti.


Subject(s)
Animals, Newborn , Organ of Corti/cytology , Animals , Base Sequence , Cell Proliferation , Cells, Cultured , DNA Primers , Kruppel-Like Factor 4 , Mice , Reverse Transcriptase Polymerase Chain Reaction
5.
BMC Cancer ; 13: 468, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24112473

ABSTRACT

BACKGROUND: Accumulating evidence has suggested the importance of glutamate signaling in cancer growth, yet the signaling pathway has not been fully elucidated. N-methyl-D-aspartic acid (NMDA) receptor activates intracellular signaling pathways such as the extracellular-signal-regulated kinase (ERK) and forkhead box, class O (FOXO). Suppression of lung carcinoma growth by NMDA receptor antagonists via the ERK pathway has been reported. However, series of evidences suggested the importance of FOXO pathways for the regulation of normal and cancer cell growth. In the liver, FOXO1 play important roles for the cell proliferation such as hepatic stellate cells as well as liver metabolism. Our aim was to investigate the involvement of the FOXO pathway and the target genes in the growth inhibitory effects of NMDA receptor antagonist MK-801 in human hepatocellular carcinoma. METHODS: Expression of NMDAR1 in cancer cell lines from different tissues was examined by Western blot. NMDA receptor subunits in HepG2, HuH-7, and HLF were examined by reverse transcriptase polymerase chain reaction (RT-PCR), and growth inhibition by MK-801 and NBQX was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The effects of MK-801 on the cell cycle were examined by flow cytometry and Western blot analysis. Expression of thioredoxin-interacting protein (TXNIP) and p27 was determined by real-time PCR and Western blotting. Activation of the FOXO pathway and TXNIP induction were examined by Western blotting, fluorescence microscopy, Chromatin immunoprecipitation (ChIP) assay, and reporter gene assay. The effects of TXNIP on growth inhibition were examined using the gene silencing technique. RESULTS: NMDA receptor subunits were expressed in all cell lines examined, and MK-801, but not NBQX, inhibited cell growth of hepatocellular carcinomas. Cell cycle analysis showed that MK-801 induced G1 cell cycle arrest by down-regulating cyclin D1 and up-regulating p27. MK-801 dephosphorylated Thr24 in FOXO1 and induced its nuclear translocation, thus increasing transcription of TXNIP, a tumor suppressor gene. Knock-down of TXNIP ameliorated the growth inhibitory effects of MK-801. CONCLUSIONS: Our results indicate that functional NMDA receptors are expressed in hepatocellular carcinomas and that the FOXO pathway is involved in the growth inhibitory effects of MK-801. This mechanism could be common in hepatocellular carcinomas examined, but other mechanisms such as ERK pathway could exist in other cancer cells as reported in lung carcinoma cells. Altered expression levels of FOXO target genes including cyclin D1 and p27 may contribute to the inhibition of G1/S cell cycle transition. Induction of the tumor suppressor gene TXNIP plays an important role in the growth inhibition by MK-801. Our report provides new evidence that FOXO-TXNIP pathway play a role in the inhibition of the hepatocellular carcinoma growth by MK-801.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carrier Proteins/metabolism , Dizocilpine Maleate/pharmacology , Forkhead Transcription Factors/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Signal Transduction , Carcinoma, Hepatocellular/genetics , Carrier Proteins/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism
6.
Biochem Biophys Res Commun ; 425(4): 717-23, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22877751

ABSTRACT

Rare sugar D-psicose has cropped up as a non-toxic and effective compound to protect and preserve pancreatic ß-islets in the growing type 2 diabetes mellitus (T2DM) rats through the regulation of glucose and fat metabolism. The present study was undertaken to examine the effect of rare sugar D-psicose on the protection of pancreatic ß-islets using Otsuka Long-Evans Tokushima Fatty (OLETF) rats, a T2DM model. Treated rats were fed with 5% D-psicose or 5% D-glucose supplemented drinking water, and only water in the control for 13 weeks. A non-diabetic Long-Evans Tokushima Otsuka (LETO), fed with water served as a counter control of OLETF. D-Psicose significantly attenuated progressive ß-islet fibrosis and preserved islets, evaluated by hematoxylin-eosin staining, Masson's trichrome staining and immunostainings of insulin and α-smooth muscle actin (SMA). D-Psicose significantly reduced increase in body weight and abdominal fat deposition. Oral glucose tolerance test (OGTT) showed reduced blood glucose levels suggesting the improvement of insulin resistance. All these data suggests that D-psicose protected and preserved pancreatic ß-islets through the maintenance of hyperglycemia and by the prevention of fat accumulation in OLETF rats.


Subject(s)
Cytoprotection , Diabetes Mellitus, Type 2/drug therapy , Fructose/administration & dosage , Insulin Resistance , Insulin-Secreting Cells/drug effects , Obesity/drug therapy , Abdominal Fat/drug effects , Abdominal Fat/pathology , Actins/metabolism , Adipokines/blood , Animals , Body Mass Index , Body Weight/drug effects , Eating/drug effects , Fibrosis , Glucose/metabolism , Glucose Tolerance Test , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Obesity/blood , Rats , Rats, Inbred OLETF
7.
Biochem Biophys Res Commun ; 405(1): 7-12, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21187061

ABSTRACT

A rare sugar, D-psicose has progressively been evaluated as a unique metabolic regulator of glucose and lipid metabolism, and thus represents a promising compound for the treatment of type 2 diabetes mellitus (T2DM). The present study was undertaken to examine the underlying effector organs of D-psicose in lowering blood glucose and abdominal fat by exploiting a T2DM rat model, Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Rats were fed 5% D-psicose or 5% D-glucose supplemented in drinking water, and only water in the control for 13 weeks and the protective effects were compared. A non-diabetic Long-Evans Tokushima Otsuka (LETO), fed with water served as a counter control of OLETF. After 13 weeks feeding, D-psicose treatment significantly reduced the increase in body weight and abdominal fat mass. Oral glucose tolerance test (OGTT) showed the reduced blood glucose and insulin levels suggesting the improvement of insulin resistance in OLETF rats. Oil-red-O staining elucidated that D-psicose significantly reduced lipid accumulation in the liver. Immunohistochemical analysis showed D-psicose induced glucokinase translocation from nucleus to cytoplasm of the liver which enhances glucokinase activity and subsequent synthesis of glycogen in the liver. D-psicose also protected the pathological change of the ß-cells of pancreatic islets. These data demonstrate that D-psicose controls blood glucose levels by reducing lipotoxicity in liver and by preserving pancreatic ß-cell function.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 2/drug therapy , Fructose/therapeutic use , Insulin Resistance , Abdominal Fat/drug effects , Abdominal Fat/pathology , Animals , Body Weight/drug effects , Cell Nucleus/enzymology , Cytoplasm/enzymology , Cytoprotection , Eating/drug effects , Fatty Liver/drug therapy , Fatty Liver/pathology , Glucokinase/metabolism , Glucose Tolerance Test , Homeostasis/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Lipid Metabolism/drug effects , Male , Pancreas/drug effects , Pancreas/pathology , Protein Transport/drug effects , Rats , Rats, Inbred OLETF
8.
Int J Oncol ; 32(2): 377-85, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202760

ABSTRACT

'Rare sugars' are defined as monosaccharides that exist in nature but are only present in limited quantities. The development of mass production method of rare sugars revealed some interesting physiological effects of these on animal cells, but the mechanisms have not been well studied. We examined the effect of D-allose on the proliferation of cancer cells and the underlying molecular mechanism of the action. The HuH-7 hepatocellular carcinoma cells were treated with various monosaccharides for 48 h and D-allose was shown to inhibit cell growth by 40% in a dose-dependent manner. D-allose induced G1 cell cycle arrest but not apoptosis. The microarray analysis revealed that D-allose significantly up-regulated thioredoxin interacting protein (TXNIP) gene expression, which is often suppressed in tumor cells and western blot analysis confirmed its increase at protein level. The overexpression of TXNIP also induced G1 cell cycle arrest. Analysis of cell cycle regulatory genes showed p27kip1, a key regulator of G1/S cell cycle transition, to be increased at the protein but not the transcriptional level. Protein interaction between TXNIP and jab1, and p27kip1 and jab1, was observed, suggesting stabilization of p27kip1 protein by the competitive inhibition of jab1-mediated nuclear export of p27kip1 by TXNIP. In addition, increased interaction and nuclear localization of TXNIP and p27kip1 were apparent after D-allose treatment. Our findings surprisingly suggest that D-allose, a simple monosaccharide, may act as a novel anticancer agent via unique TXNIP induction and p27kip1 protein stabilization.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carrier Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , G1 Phase , Gene Expression Regulation, Neoplastic , Glucose/pharmacology , Liver Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Models, Biological , Reverse Transcriptase Polymerase Chain Reaction
9.
Hear Res ; 365: 110-126, 2018 08.
Article in English | MEDLINE | ID: mdl-29804721

ABSTRACT

The cochlea contains macrophages. These cells participate in inflammatory responses to cochlear pathogenesis. However, it is not clear how and when these cells populate the cochlea during postnatal development. The current study aims to determine the postnatal development of cochlear macrophages with the focus on macrophage development in the organ of Corti and the basilar membrane. Cochleae were collected from C57BL/6J mice at ages of postnatal day (P) 1 to P21, as well as from mature mice (1-4 months). Macrophages were identified based on their expression of F4/80 and Iba1, as well as their unique morphologies. Two sets of macrophages were identified in the regions of the organ of Corti and the basilar membrane. One set resides on the scala tympani side of the basilar membrane. These cells have a round shape at P1 and start to undergo site-specific differentiation at P4. Apical macrophages adopt a dendritic shape. Middle and basal macrophages take on an irregular shape with short projections. Basal macrophages further differentiate into an amoeboid shape. The other set of macrophages resides above the basilar membrane, either beneath the cells of the organ of Corti or along the spiral vessel of the basilar membrane. As the sensory epithelium matures, these cells undergo developmental death with the phenotypes of apoptosis. Macrophages are also identified in the spiral ligament, spiral limbus, and neural regions. Their numbers decrease during postnatal development. Together, these results suggest a dynamic rearrangement of the macrophage population during postnatal cochlear development.


Subject(s)
Cell Differentiation , Cochlea/physiology , Macrophages/physiology , Age Factors , Animals , Animals, Newborn , Antigens, Differentiation/metabolism , Apoptosis , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Cell Shape , Cochlea/metabolism , Cochlea/ultrastructure , Female , Leukocyte Common Antigens/metabolism , Macrophages/metabolism , Macrophages/ultrastructure , Male , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Phenotype
10.
FEBS Open Bio ; 8(11): 1804-1819, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30410860

ABSTRACT

Thioredoxin interacting protein (TXNIP) is a novel tumor suppressor that is down-regulated in several cancer tissues and tumor cell lines. Overexpression of TXNIP causes cell cycle arrest at the G1/S checkpoint in the hepatocellular carcinoma cell line HuH-7. TXNIP contains putative phosphorylation sites, but the effects of its phosphorylation have not been fully characterized. TXNIP also contains two α-arrestin domains (N-arrestin and C-arrestin) whose functions are not fully understood. Here, we reveal an association between TXNIP and cell cycle regulatory proteins (p27kip1, Jun activation domain-binding protein 1 (JAB1), Cdk2, and cyclin E), suggesting its participation in cell cycle regulation. We observed phosphorylation of TXNIP and used both in vivo and in vitro kinase assays to demonstrate that TXNIP can be phosphorylated by p38 mitogen-activated protein kinase. Furthermore, we also identified Ser361 in TXNIP as one of the major phosphorylation sites. Cell cycle analysis showed that Ser361 phosphorylation participates in TXNIP-mediated cell cycle arrest. In addition, the C-arrestin domain may also play an important role in cell cycle arrest. We also showed that phosphorylation at Ser361 may be important for the association of TXNIP with JAB1 and that the C-arrestin domain is necessary for the nuclear localization of this molecule. Collectively, these studies reveal that TXNIP participates in cell cycle regulation through association with regulatory proteins, especially JAB1, and that C-arrestin-dependent nuclear localization is important for this function. This work may facilitate the development of a new cancer therapy strategy that targets TXNIP as a key molecule inhibiting cancer cell growth via cell cycle blockade at the G1/S checkpoint.

11.
Oncol Rep ; 15(4): 765-71, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16525656

ABSTRACT

We have previously demonstrated the inverse correlation of Jab1 and p27 proteins, as well as prognostic significance in epithelial ovarian carcinomas. In order to investigate Skp2 protein and its correlation with Jab1, p27, and clinical outcome, we evaluated Skp2 expression in a group of epithelial ovarian tumors. Immunohistochemical analysis was performed on 80 cases of ovarian tumors (33 benign and 47 malignant), and 26 of the 80 cases were evaluated by Western blot analysis. Immunofluorescence was carried out in the human ovarian adenocarcinoma cell line OVCAR-3. Skp2 expression was detected in 53.2% of malignant tumors and 18.2% of benign tumors. The positive ratio of Skp2 expression was increased from benign to malignant ovarian tumors (p=0.002). A negative correlation between Skp2 and p27 was found in benign and malignant ovarian tumors (p=0.006 and p<0.0001, respectively). Skp2 expression was significantly associated with high tumor grade (p=0.001), lymph node metastasis (p=0.01), and residual disease (p=0.012). Kaplan-Meier survival analysis showed that Skp2 expression was significantly associated with poor prognosis (p=0.013), and patients with Skp2(+)/Jab1(+)p27(-) expression had the worst prognosis among all phenotypes of Skp2/Jab1/p27 expression (p=0.0007). Our results suggest that Skp2 expression was significantly associated with malignancy, and the Skp2 protein level may be a valuable prognostic factor for epithelial ovarian carcinomas. Furthermore, the combined evaluation of Skp2/Jab1/p27 proteins provides important prognostic information on patients with epithelial ovarian carcinoma.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Ovarian Neoplasms/pathology , Peptide Hydrolases/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Blotting, Western , COP9 Signalosome Complex , Cell Line, Tumor , Epithelial Cells/chemistry , Epithelial Cells/pathology , Female , Humans , Immunohistochemistry , Middle Aged , Ovarian Neoplasms/metabolism , Prognosis , Survival Analysis
12.
Oncol Rep ; 15(4): 773-8, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16525657

ABSTRACT

The tumor suppressor PTEN, phosphatase and tensin homolog on chromosome 10, plays an essential role in regulating signaling pathways involved in cell growth and apoptosis and is inactivated in a wide variety of tumors. Survivin, a member of the inhibitor of apoptosis protein family (IAP), is associated with cell proliferation, and overexpressed in common human tumors. Both PTEN and survivin proteins can regulate cell cycle and apoptosis, but their biological effects are adverse. We have previously investigated the role of survivin expression in epithelial ovarian tumors. In this study, we evaluated the alteration and clinical relevance of PTEN expression and further assessed its correlation with survivin expression in epithelial ovarian tumors. Immunohistochemical analysis was performed in 103 cases of ovarian tumors, and 26 of the 103 cases were evaluated by Western blot analysis. PTEN expression was reduced from benign to malignant ovarian tumors (p=0.0003), and an inverse correlation between PTEN and survivin was found in benign, borderline, and malignant tumors (p=0.004, p=0.015 and p=0.0005, respectively). PTEN expression was significantly associated with tumor grade (p=0.001), histological subtype (p=0.037), ascites (p=0.038), and residual disease (p=0.0006). Kaplan-Meier survival analysis showed that the loss of PTEN expression was significantly associated with poor overall survival (p=0.021), and patients with PTEN(-)/survivin(+) expression had the worst prognosis among all phenotypes of PTEN/survivin expression (p=0.039). Our results suggest that the altered PTEN expression and its inverse correlation with survivin may be involved in the development and progression of ovarian tumors, and the combined detection of PTEN and survivin proteins might be more valuable in the evaluation of malignancy and prognosis in epithelial ovarian tumors.


Subject(s)
Microtubule-Associated Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Ovarian Neoplasms/pathology , PTEN Phosphohydrolase/biosynthesis , Epithelial Cells/chemistry , Epithelial Cells/pathology , Female , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Middle Aged , Ovarian Neoplasms/metabolism , Prognosis , Survival Analysis , Survivin
13.
Clin Cancer Res ; 11(1): 259-66, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15671554

ABSTRACT

PURPOSE: Jun activation domain-binding protein 1 (Jab1) is known as a coactivator of AP1 transcription factor, which contributes to tumor progression by degrading the p27kip1 protein. The purpose of this study is to investigate whether Jab1 expression is correlated with p27kip1 level and cell proliferation, as well as whether Jab1 expression is associated with clinicopathologic variables and prognosis of laryngeal squamous cell carcinoma (LSCC). EXPERIMENTAL DESIGN: Immunohistochemical and/or Western blot analysis was done in HEp-2 cells and 102 cases of LSCCs. RESULTS: Jab1 expression was negatively associated with p27kip1 expression and was positively associated with cell proliferation both in HEp-2 cells and LSCCs. Jab1 overexpression was detected in 51% LSCCs and was significantly associated with unfavorable clinicopathologic variables. Survival analysis revealed that Jab1 overexpression is significantly associated with short disease-free and overall survival (P = 0.0036 and P = 0.0001, respectively). When Jab1 and p27kip1 are combined, patients with Jab1(+)/p27kip1(-) revealed poor disease-free and overall survival (P= 0.0008 and P < 0.0001, respectively). When Jab1 expression and lymph node status are combined, patients with Jab1(+)/lymph node(+) revealed poorer disease-free andoverall survival than others (P < 0.0001 and P < 0.0001, respectively). Furthermore, patients with the phenotype of Jab1(+)/p27kip1(-)/lymph node(+) revealed the worst disease-free and overall survival (P < 0.0001 and P < 0.0001, respectively). Multivariate analysis revealed that Jab1 protein is an independent prognostic indicator for overall survival. CONCLUSIONS: These findings suggested that Jab1 protein may contribute to the tumor progression and represent a novel prognostic indicator for LSCCs.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/mortality , Peptide Hydrolases/biosynthesis , Prognosis , Transcription Factors/biosynthesis , Adult , Animals , Blotting, Western , COP9 Signalosome Complex , Carcinoma, Squamous Cell/diagnosis , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27 , Disease Progression , Disease-Free Survival , Female , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Laryngeal Neoplasms/diagnosis , Lymphatic Metastasis , Male , Mice , Microscopy, Fluorescence , Middle Aged , Models, Statistical , Multivariate Analysis , NIH 3T3 Cells , Phenotype , Proportional Hazards Models , Retrospective Studies , Time Factors , Treatment Outcome , Tumor Suppressor Proteins/biosynthesis
14.
Heliyon ; 2(1): e00055, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27441238

ABSTRACT

Although various parts of J. curcas (Jatropha curcas L., Euphorbiaceae) have long been used as traditional folk medicines for their antiviral, analgesic, and/or antidotal efficacies, we are the first to investigate the role of anti-carcinogenicity of isoamericanol A (IAA) from the seed extract. Our results showed that IAA is capable of inhibiting cell proliferation in a dose-dependent manner on the human cancer cell lines of MCF-7, MDA-MB231, HuH-7, and HeLa. Flow cytometry analysis showed IAA significantly induces cell cycle arrest at G2/M on MCF-7 cells. At both protein and mRNA levels examined by western blot and real-time PCR, the results revealed increased expression of BTG2 (B-cell translocation gene 2), p21 (p21(WAF1/CIPI) ), and GADD45A (growth arrest and DNA-damage-inducible, alpha) after IAA treatment, but inversed expression in CDK1 (cyclin-dependent kinase 1) and cyclins B1 and B2. All these effects contribute to G2/M cell cycle arrest. Furthermore, these results coincide with the changes in molecular expressions determined by DNA-microarray analysis. Our findings indicate that IAA has an inhibitory effect on cell proliferation of MCF-7 through cell cycle arrest, giving it great potential as a future therapeutic reagent for cancers.

15.
Int J Oncol ; 27(4): 907-12, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142305

ABSTRACT

Rare sugars are monosaccharides distributed rarely in nature, because of its very limited amount and cost, the biological effect has hardly been studied. Recently, an effective strategy for mass production of rare sugars has been developed. As a result, a wide range of study of rare sugars from the basic to applied research has become possible. For biological application of rare sugars, it is necessary to fundamentally investigate the relationships between rare sugars and living cells in terms of physiology. Therefore, we firstly examined the effect of rare sugars including D-psicose, D-allose, D-altrose and D-talitol on cell proliferation using certain cell lines in vitro. Cell growth was evaluated by MTT assay after 24-, 48- and 72-h treatment. The result shows that D-allose has a significant inhibitory effect on cancer cell proliferation in a dose-dependent manner. Although the exact mechanism remains unclear, this finding represents a novel aspect of the biological profile of D-allose and suggests that D-allose may be an effective adjuvant therapeutic agent against cancer in the future.


Subject(s)
Gene Expression Regulation, Neoplastic , Glucose/chemistry , Glucose/pharmacology , Neoplasms/drug therapy , Apoptosis , Blotting, Western , Cell Cycle , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/chemistry , Fructose/pharmacology , HeLa Cells , Hexoses/pharmacology , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Models, Chemical , Monosaccharides/chemistry , Reactive Oxygen Species , Sugar Alcohols/pharmacology , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors
16.
Anticancer Res ; 25(4): 2639-44, 2005.
Article in English | MEDLINE | ID: mdl-16080505

ABSTRACT

BACKGROUND: D-allose is a rare sugar found in nature and, because of its very limited amount and of the high cost associated with its synthesis, its physiological functions remain virtually unknown. The aim of the current study was to investigate the effect of D-allose on the proliferation of human ovarian carcinoma cells in vitro. MATERIALS AND METHODS: Human ovarian carcinoma cells (OVCAR-3 cell line) were exposed to rare sugars including D-allose, D-altrose, D-psicose and D-talitol. Cell growth was evaluated by MTT assay. Cell cycle analysis was carried out by flow cytometric assay. The expression of cell cycle regulatory proteins was determined by Western blot analysis. TUNEL assay was employed for the detection of apoptotic cells. RESULTS: D-allose had a significant inhibitory effect on ovarian cancer cell proliferation in a dose-dependent manner, and caused a moderate G2/M arrest in the cell cycle, up-regulation of Cdk inhibitors p21 and p27 levels, and the induction of apoptosis in OVCAR-3 cells. CONCLUSION: Our results show, for the first time, that D-allose inhibits the growth of ovarian carcinoma cells in vitro. Although the exact mechanisms remain unclear, these findings suggest that D-allose possesses a novel inhibitory property on ovarian carcinoma cell proliferation, and may represent a new class of compounds with possible therapeutic potential.


Subject(s)
Adenocarcinoma/drug therapy , Glucose/pharmacology , Ovarian Neoplasms/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle Proteins/biosynthesis , Cell Growth Processes/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Dose-Response Relationship, Drug , Female , Fructose/pharmacology , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Sugar Alcohols/pharmacology , Tumor Suppressor Proteins/biosynthesis
17.
Drug Des Devel Ther ; 9: 525-35, 2015.
Article in English | MEDLINE | ID: mdl-25632221

ABSTRACT

BACKGROUND: The fundamental cause of overweight and obesity is consumption of calorie-dense foods. We have introduced a zero-calorie sweet sugar, d-psicose (d-allulose), a rare sugar that has been proven to have strong antihyperglycemic and antihyperlipidemic effects, and could be used as a replacement of natural sugar for the obese and diabetic subjects. AIM: Above mentioned efficacy of d-psicose (d-allulose) has been confirmed in our previous studies on type 2 diabetes mellitus (T2DM) model Otsuka Long-Evans Tokushima Fatty (OLETF) rats with short-term treatment. In this study we investigated the long-term effect of d-psicose in preventing the commencement and progression of T2DM with the mechanism of preservation of pancreatic ß-cells in OLETF rats. METHODS: Treated OLETF rats were fed 5% d-psicose dissolved in water and control rats only water. Nondiabetic control rats, Long-Evans Tokushima Otsuka (LETO), were taken as healthy control and fed water. To follow the progression of diabetes, periodic measurements of blood glucose, plasma insulin, and body weight changes were continued till sacrifice at 60 weeks. Periodic in vivo body fat mass was measured. On sacrifice, pancreas, liver, and abdominal adipose tissues were collected for various staining tests. RESULTS: d-Psicose prevented the commencement and progression of T2DM till 60 weeks through the maintenance of blood glucose levels, decrease in body weight gain, and the control of postprandial hyperglycemia, with decreased levels of HbA1c in comparison to nontreated control rats. This improvement in glycemic control was accompanied by the maintenance of plasma insulin levels and the preservation of pancreatic ß-cells with the significant reduction in inflammatory markers. Body fat accumulation was significantly lower in the treatment group, with decreased infiltration of macrophages in the abdominal adipose tissue. CONCLUSION: Our findings suggest that the rare sugar d-psicose could be beneficial for the prevention and control of obesity and hyperglycemia with the preservation of ß-cells in the progression of T2DM.


Subject(s)
Diabetes Mellitus, Type 2/prevention & control , Fructose/pharmacology , Hypoglycemic Agents/pharmacology , Abdominal Fat/drug effects , Abdominal Fat/metabolism , Abdominal Fat/physiopathology , Adiposity/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Obesity Agents/pharmacology , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Disease Progression , Glycated Hemoglobin/metabolism , Inflammation/blood , Inflammation/physiopathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Insulin/blood , Insulin Resistance , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Obesity/blood , Obesity/physiopathology , Obesity/prevention & control , Rats, Inbred OLETF , Time Factors , Weight Gain/drug effects
18.
Cancer Lett ; 177(1): 13-9, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11809526

ABSTRACT

Cyclin B1 is a key molecule for G2/M phase transition during the cell cycle and is overexpressed in various human tumors. However, the expression status of cyclin B1 in laryngeal squamous cell carcinoma (LSCC) and its clinical significance remain unknown. We used immunohistochemical studies to examine the expression of cyclin B1 in 102 patients with LSCC. The results showed that cyclin B1 overexpression was observed in 40 cases (39.2%) of LSCCs and was significantly correlated with the tumor site (P=0.031), tumor size (P<0.0001), and advanced stage (P=0.003). In addition, cyclin B1 overexpression was associated with patients' overall survival, but not with disease-free survival using Kaplan-Meier analysis. On multivariate analysis, cyclin B1 expression was not recognized as an independent prognostic factor. These findings indicate that cyclin B1 overexpression may be associated with the malignant biological behavior of LSCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cyclin B/metabolism , Laryngeal Neoplasms/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Cyclin B1 , Disease-Free Survival , Female , Humans , Immunoenzyme Techniques , Laryngeal Neoplasms/pathology , Laryngeal Neoplasms/therapy , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Prognosis , Proliferating Cell Nuclear Antigen/metabolism , Survival Rate
19.
Int J Oncol ; 21(2): 315-20, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12118327

ABSTRACT

Survivin is a new member of the inhibitors of apoptosis proteins (IAP) family, selectively overexpressed in common human cancers but not in normal adult tissues, and associated with aggressiveness of the disease and unfavorable outcomes. Recent study also found that survivin expression is associated with cell proliferation. In order to gain insight into the role of survivin in ovarian tumors, we investigated the expression of survivin in a group of epithelial ovarian tumors, and examined the relationship of its expression with cell proliferation and clinical outcome. Immunohistochemical analysis was performed in 103 cases of epithelial ovarian tumors. Twenty-six of the 103 cases were evaluated by Western blot analysis. The results showed that survivin overexpression was detected in 21.2% (7 of 33) of benign tumors, 47.8% (11 of 23) of borderline tumors, and 51.1% (24 of 47) of ovarian carcinomas. The positive ratio was significantly higher in malignant or borderline tumors than in benign tumors, and the overexpression of survivin was significantly correlated with the size of residual disease. A positive correlation between survivin expression and proliferative activity of tumor cell measured by PCNA index was found. Kaplan-Meier analysis demonstrated that the patients with survivin overexpression have a short overall survival. These findings suggest that survivin overexpression may play a pivotal role in the progression of ovarian tumors and may provide an important prognostic implication for epithelial ovarian carcinomas.


Subject(s)
Cysteine Proteinase Inhibitors/metabolism , Microtubule-Associated Proteins/metabolism , Ovarian Neoplasms/metabolism , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/pathology , Adolescent , Adult , Aged , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Case-Control Studies , Cell Differentiation , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Immunoenzyme Techniques , Inhibitor of Apoptosis Proteins , Lymphatic Metastasis , Middle Aged , Neoplasm Proteins , Neoplasm Staging , Ovarian Neoplasms/pathology , Prognosis , Proliferating Cell Nuclear Antigen/metabolism , Survival Rate , Survivin
20.
Neuroreport ; 14(5): 759-61, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12692478

ABSTRACT

The F-box protein, Skp2 positively regulates the G1-S transition by controlling the stability of several G1 regulators, including p27. To evaluate the role of Skp2 in the mammalian developing auditory systems, the expression of Skp2 was examined together with the expression of p27 in auditory systems. Our data show expression of Skp2 in auditory epithelia and neurons at an early stage of development. During differentiation processes, the onset of p27 expression was observed together with the down-regulation of Skp2 expression, in auditory epithelia. In contrast, an alteration of expression of p27 and Skp2 in the greater epithelial ridge and spiral ganglion appeared after differentiation of hair cells. These findings suggest that Skp2 plays a crucial role in development of mammalian auditory systems.


Subject(s)
Cell Cycle Proteins/metabolism , Cochlea/embryology , Cochlea/metabolism , Animals , Cell Differentiation/physiology , Cell Division/physiology , Cochlea/cytology , Cyclin-Dependent Kinase Inhibitor p27 , Dyneins , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Gestational Age , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Ki-67 Antigen/biosynthesis , Mice , Mice, Inbred ICR , Myosin VIIa , Myosins/biosynthesis , Neurons/cytology , Neurons/metabolism , S-Phase Kinase-Associated Proteins , Spiral Ganglion/cytology , Spiral Ganglion/embryology , Spiral Ganglion/metabolism , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL