Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
World J Gastroenterol ; 29(34): 5075-5081, 2023 Sep 14.
Article in English | MEDLINE | ID: mdl-37753365

ABSTRACT

BACKGROUND: Primary biliary cholangitis (PBC) is a chronic progressive autoimmune cholestatic disease. The main target organ of PBC is the liver, and nonsuppurative inflammation of the small intrahepatic bile ducts may eventually develop into cirrhosis or liver fibrosis. AIM: To explore the clinical characteristics of early-stage PBC, identify PBC in the early clinical stage, and promptly treat and monitor PBC. METHODS: The data of 82 patients with PBC confirmed by pathology at Tianjin Second People's Hospital from January 2013 to November 2021 were collected, and the patients were divided into stage I, stage II, stage III, and stage IV according to the pathological stage. The general data, serum biochemistry, immunoglobulins, and autoimmune antibodies of patients in each stage were retrospectively analyzed. RESULTS: In early-stage (stages I + II) PBC patients, 50.0% of patients had normal alanine aminotransferase (ALT) levels, and 37.5% had normal aspartate aminotransferase (AST) levels. For the remaining patients, the ALT and AST levels were mildly elevated; all of these patients had levels of < 3 times the upper limit of normal values. The AST levels were significantly different among the three groups (stages I + II vs stage III vs stage IV, P < 0.05). In the early stage, 29.2% of patients had normal alkaline phosphatase (ALP) levels. The remaining patients had different degrees of ALP elevation; 6.3% had ALP levels > 5 times the upper limit of normal value. Moreover, γ-glutamyl transferase (GGT) was more robustly elevated, as 29.2% of patients had GGT levels of > 10 times the upper limit of normal value. The ALP values among the three groups were significantly different (P < 0.05). In early stage, the jaundice index did not increase significantly, but it gradually increased with disease progression. However, the above indicators were significantly different (P < 0.05) between the early-stage group and the stage IV group. With the progression of the disease, the levels of albumin and albumin/globulin ratio tended to decrease, and the difference among the three groups was statistically significant (P < 0.05). In early-stage patients, IgM and IgG levels as well as cholesterol levels were mildly elevated, but there were no significant differences among the three groups. Triglyceride levels were normal in the early-stage group, and the differences among the three groups were statistically significant (P < 0.05). The early detection rates of anti-mitochondria antibody (AMA) and AMA-M2 were 66.7% and 45.8%, respectively. The positive rate of anti-sp100 antibodies was significantly higher in patients with stage IV PBC. When AMA and AMA-M2 were negative, in the early stage, the highest autoantibody was anti-nuclear antibody (ANA) (92.3%), and in all ANA patterns, the highest was ANA centromere (38.5%). CONCLUSION: In early-stage PBC patients, ALT and AST levels are normal or mildly elevated, GGT and ALP levels are not elevated in parallel, GGT levels are more robustly elevated, and ALP levels are normal in some patients. When AMA and AMA-M2 are negative, ANA especially ANA centromere positivity suggests the possibility of early PBC. Therefore, in the clinic, significantly elevated GGT levels with or without normal ALP levels and with ANA (particularly ANA centromere) positivity (when AMA and AMA-M2 are negative) may indicate the possibility of early PBC.


Subject(s)
Autoimmune Diseases , Liver Cirrhosis, Biliary , Humans , Autoantibodies , Liver Cirrhosis, Biliary/diagnosis , Retrospective Studies , Albumins , Biomarkers
2.
Cardiovasc Toxicol ; 21(9): 710-720, 2021 09.
Article in English | MEDLINE | ID: mdl-34037971

ABSTRACT

This study aimed to discover the effect of miR-124/STAT3 axis on the inflammation and cell apoptosis in myocardial infarction (MI) rats. Sprague-Dawley (SD) male rats were selected for establishing MI models and divided into Sham, MI, MI + anti-miR-124 and MI + Ad-miR-124 groups. Cardiac function was detected via echocardiography. Hematoxylin & eosin (HE) and triphenyltetrazolium chloride (TTC) staining were used to observe the pathological changes and infarction area, while transferase (TdT)-mediated D-UTP-biotin nick end labeling (TUNEL) assay was to observe myocardial apoptosis. Enzyme-linked immunosorbent assay (ELISA) was used to determine the serum levels of inflammatory cytokines. Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and western blotting were performed to determine the mRNA and protein levels, respectively. Dual luciferase reporter gene assay revealed that STAT3 was a target gene of miR-124. The expression levels of miR-124 were increased and the pSTAT3/STAT3 ratio was reduced in the MI rats. The rats in the MI group showed enhanced LVEDD and LVESD, reduced LVEF and LVFS, as well as larger myocardial infarction area compared with the Sham group, Besides, IL-1ß, IL-6, TNF-α and MCP-1 levels were elevated and the expressions of Bax/Bcl-2 ratio and cleaved caspase-3 were downregulated in MI group. We further found that silencing miR-124 improved cardiac function, reduced infarction area and the levels of inflammatory cytokines, as well as prevented myocardial apoptosis in MI rats. Silencing miR-124 could inhibit the inflammation and apoptosis of myocardial cells, thereby relieving the MI injury via upregulation of STAT3.


Subject(s)
Apoptosis , Cytokines/metabolism , Inflammation Mediators/metabolism , MicroRNAs/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , STAT3 Transcription Factor/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cytokines/genetics , Disease Models, Animal , Gene Expression Regulation , Male , MicroRNAs/genetics , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Phosphorylation , Rats, Sprague-Dawley , STAT3 Transcription Factor/genetics , Signal Transduction , Stroke Volume , Ventricular Function, Left
3.
Cancer Commun (Lond) ; 41(3): 258-272, 2021 03.
Article in English | MEDLINE | ID: mdl-33570246

ABSTRACT

BACKGROUND: Insulin gene enhancer protein 1, (ISL1), a LIM-homeodomain transcription factor, is involved in multiple tumors and is associated with insulin secretion and metabolic phenotypes. However, the role of ISL1 in stimulating glycolysis to promote tumorigenesis in gastric cancer (GC) is unclear. In this study, we aimed to characterize the expression pattern of ISL1 in GC patients and explore its molecular biological mechanism in glycolysis and tumorigenesis. METHODS: We analyzed the expression and clinical significance of ISL1 in GC using immunohistochemistry and real-time polymerase chain reaction (PCR). Flow cytometry and IncuCyte assays were used to measure cell proliferation after ISL1 knockdown. RNA-sequencing was performed to identify differentially expressed genes, followed by Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and Gene Set Enrichment Analysis (GSEA) to reveal key signaling pathways likely regulated by ISL1 in GC. Alteration of the glycolytic ability of GC cells with ISL1 knockdown was validated by measuring the extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) and by detecting glucose consumption and lactate production. The expression of glucose transporter 4 (GLUT4) and ISL1 was assessed by Western blotting, immunohistochemistry, and immunofluorescent microscopy. The luciferase reporter activity and chromatin immunoprecipitation assays were performed to determine the transcriptional regulation of ISL1 on GLUT4. RESULTS: High levels of ISL1 and GLUT4 expression was associated with short survival of GC patients. ISL1 knockdown inhibited cell proliferation both in vitro and in vivo. KEGG analysis and GSEA for RNA-sequencing data indicated impairment of the glycolysis pathway in GC cells with ISL1 knockdown, which was validated by reduced glucose uptake and lactate production, decreased ECAR, and increased OCR. Mechanistic investigation indicated that ISL1 transcriptionally regulated GLUT4 through binding to its promoter. CONCLUSION: ISL1 facilitates glycolysis and tumorigenesis in GC via the transcriptional regulation of GLUT4.


Subject(s)
Insulins , Stomach Neoplasms , Carcinogenesis , Cell Line, Tumor , Glucose Transport Proteins, Facilitative , Glycolysis/genetics , Humans , Stomach Neoplasms/genetics
4.
J Cell Mol Med ; 24(21): 12355-12367, 2020 11.
Article in English | MEDLINE | ID: mdl-32961025

ABSTRACT

Diabetes is a disorder of glucose metabolism, and over 90% are type 2 diabetes. Diabetic cardiomyopathy (DCM) is one of the type 2 diabetes complications, usually accompanied by changes in myocardial structure and function, together with cardiomyocyte apoptosis. Our study investigated the effect of curcumin on regulating oxidative stress (OS) and apoptosis in DCM. In vivo, diabetes was induced in an experimental rat model by streptozoticin (STZ) together with high-glucose and high-fat (HG/HF) diet feeding. In vitro, H9c2 cardiomyocytes were cultured with high-glucose and saturated free fatty acid palmitate. Curcumin was orally or directly administered to rats or cells, respectively. Streptozoticin -induced diabetic rats showed metabolism abnormalities and elevated markers of OS (superoxide dismutase [SOD], malondialdehyde [MDA], gp91phox , Cyt-Cyto C), enhanced cell apoptosis (Bax/Bcl-2, Cleaved caspase-3, TUNEL-positive cells), together with reduced Akt phosphorylation and increased Foxo1 acetylation. Curcumin attenuated the myocardial dysfunction, OS and apoptosis in the heart of diabetic rats. Curcumin treatment also enhanced phosphorylation of Akt and inhibited acetylation of Foxo1. These results strongly suggest that apoptosis was increased in the heart of diabetic rats, and curcumin played a role in diabetic cardiomyopathy treatment by modulating the Sirt1-Foxo1 and PI3K-Akt pathways.


Subject(s)
Apoptosis/drug effects , Curcumin/pharmacology , Diabetic Cardiomyopathies/drug therapy , Oxidative Stress/drug effects , Signal Transduction/drug effects , Animals , Blood Glucose/metabolism , Cell Survival , Diabetes Mellitus, Experimental , Male , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Sirtuin 1/metabolism
5.
Eur J Med Chem ; 187: 111982, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31877538

ABSTRACT

Alzheimer's disease (AD) is characterized by progressive decrease in cognitive abilities, language impairment and irreversible memory loss. Amyloid-ß (Aß) and tau deposits are essential as the major factors involved in the pathogenesis of AD. Unfortunately, anti-Alzheimer's disease agents currently available are not potent enough to reverse the cause of the disease. Interestingly, near infrared fluorescence (NIRF) dyes have been marked as promising tools in analytical researches and are often adopted as molecular probes to monitor and diagnose AD, in vitro or in vivo. Compared with other imaging techniques, such as positron emission tomography (PET), magnetic resonance imaging (MRI), and single-photon emission computed tomography (SPECT), NIRF dye probes have been applied in AD pre-clinical trials and have gained rapid benefits, in view of their advantages including real time imaging, biocompatibility, high selectivity and sensitivity, high spatiotemporal resolution, and easy data analysis. This work reviews the developmental design of typical NIRF dye probes in monitoring Aßs and tau species in the brain of AD model mice and patients.


Subject(s)
Alzheimer Disease/diagnostic imaging , Fluorescent Dyes/chemistry , Animals , Humans , Infrared Rays
6.
Cancer Manag Res ; 11: 5375-5391, 2019.
Article in English | MEDLINE | ID: mdl-31354346

ABSTRACT

Purpose: Emerging evidence has shown that long noncoding RNAs (lncRNAs) participate in oncogenesis and tumor progression. We previously found a novel lncRNA p4516 which was closely associated with prognosis by preliminary study of lncRNA expression profile from paired tumors and nontumor tissues in 198 gastric cancer (GC) patients. However, the exact biological functions and the underlying molecular mechanisms of p4516 in gastric tumorigenesis still remain unclear. Materials and methods: The RNA fluorescence in situ hybridization (RNA-FISH) analysis, cytoplasmic and nuclear RNA isolation and qRT-PCR were applied to determine the subcellular localization of p4516. Expression levels of p4516 were assessed using qRT-PCR in both GC cell lines and in 142 primary GC tissues. Correlations between p4516 expression and GC patients' clinicopathological parameters were analyzed. Gain- and loss-of-function experiments were employed to investigate the role of p4516 in proliferation, migration and invasion both in vitro and in vivo. In addition, Western blotting and immunohistochemical staining were used to examine the protein expression levels. Results: LncRNA p4516 was mainly localized in the nucleus of GC cells and p4516 tended to have higher expression levels in GC cells compared to the normal gastric mucosa-derived cells GES-1. Furthermore, higher expression levels of p4516 correlated with worse clinical outcomes in GC patients and acted as an independent prognostic biomarker. Functional analysis revealed that p4516 participated in the regulation of GC cell proliferation, invasion and migration both in vivo and in vitro. Moreover, p4516 was involved in epithelial-mesenchymal transition (EMT) in GC cells. Conclusion: Our study demonstrated the oncogenic role of novel lncRNA p4516 in the gastric carcinogenesis for the first time. High expression of p4516 may act as prognostic marker in patient with gastric cancer.

7.
ACS Chem Neurosci ; 10(8): 3500-3509, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31244052

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder, characterized by progressive loss of memory and cognitive function, and is associated with the deficiency of synaptic acetylcholine, as well as chronic neuroinflmmation. Tacrine, a potent acetylcholinesterase (AChE) inhibitor, was previously a prescribed clinical therapeutic agent for AD, but it was recently withdrawn because it caused widespread hepatotoxicity. Hydrogen sulfide (H2S) has neuroprotective, hepatoprotective, and anti-inflammatory effects. In this study, we synthesized a new compound, a tacrine-H2S donor hybrid (THS) by introducing H2S-releasing moieties (ACS81) to tacrine. Subsequently, pharmacological and biological evaluations of THS were conducted in the aluminum trichloride (AlCl3)-induced AD mice model. We found that THS (15 mmol/kg) improved cognitive and locomotor activity in AD mice in the step-through test and open field test, respectively. THS showed strong AChE inhibitory activity in the serum and hippocampus of AD mice and induced increased hippocampal H2S levels. Furthermore, THS reduced mRNA expression of the proinflammatory cytokines, TNF-α, IL-6, and IL-1ß and increased synapse-associated proteins (synaptophysin and postsynaptic density protein 95) in the hippocampus of AD mice. Importantly, THS, unlike tacrine, did not increase liver transaminases (alanine transaminase and aspartate transaminase) or proinflammatory cytokines, indicating THS is much safer than tacrine. Therefore, the multifunctional effects of this new hybrid compound of tacrine and H2S indicate it is a promising compound for further research into the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Cognition/drug effects , Cognitive Dysfunction/drug therapy , Hydrogen Sulfide/therapeutic use , Neuroprotective Agents/therapeutic use , Tacrine/therapeutic use , Acetylcholinesterase/metabolism , Aluminum Chloride , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Animals , Cholinesterase Inhibitors/pharmacology , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Cytokines/metabolism , Disease Models, Animal , Drug Combinations , Hippocampus/drug effects , Hippocampus/metabolism , Hydrogen Sulfide/pharmacology , Male , Memory/drug effects , Mice , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Synapses/drug effects , Synapses/metabolism , Tacrine/pharmacology
8.
Cell Death Dis ; 10(2): 33, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30674889

ABSTRACT

ISL1, a LIM-homeodomain transcription factor, serves as a biomarker of metastasis in multiple tumors. However, the function and underlying mechanisms of ISL1 in gastric cancer (GC) have not been fully elucidated. Here we found that ISL1 was frequently overexpressed in GC FFPE samples (104/196, 53.06%), and associated with worse clinical outcomes. Furthermore, the overexpression of ISL1 and loss-of-function of ISL1 influenced cell proliferation, invasion and migration in vitro and in vivo, including GC patient-derived xenograft models. We used ChIP-seq and RNA-seq to identify that ISL1 influenced the regulation of H3K4 methylation and bound to ZEB1, a key regulator of the epithelial-mesenchymal transition (EMT). Meanwhile, we validated ISL1 as activating ZEB1 promoter through influencing H3K4me3. We confirmed that a complex between ISL1 and SETD7 (a histone H3K4-specific methyltransferase) can directly bind to the ZEB1 promoter to activate its expression in GC cells by immunoprecipitation, mass spectrometry, and ChIP-re-ChIP. Moreover, ZEB1 expression was significantly positively correlated with ISL1 and was positively associated with a worse outcome in primary GC specimens. Our paper uncovers a molecular mechanism of ISL1 promoting metastasis of GC through binding to the ZEB1 promoter together with co-factor SETD7. ISL1 might be a potential prognostic biomarker of GC.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , LIM-Homeodomain Proteins/biosynthesis , Stomach Neoplasms/genetics , Transcription Factors/biosynthesis , Zinc Finger E-box-Binding Homeobox 1/genetics , Animals , Cell Line, Tumor , Disease Progression , Female , HEK293 Cells , Heterografts , Humans , LIM-Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcription Factors/genetics
9.
Mar Drugs ; 17(1)2019 Jan 03.
Article in English | MEDLINE | ID: mdl-30609815

ABSTRACT

Increasing evidence indicates that environmental tobacco smoke (ETS) impairs cognitive function and induces oxidative stress in the brain. Recently, astaxanthin (ATX), a marine bioactive compound, has been reported to ameliorate cognitive deficits. However, the underlying pathogenesis remains unclear. In this study, ATX administration (40 mg/kg and 80 mg/kg, oral gavage) and cigarette smoking were carried out once a day for 10 weeks to investigate whether the p38 MAPK is involved in cognitive function in response to ATX treatment in the cortex and hippocampus of ETS mice. Results indicated that ATX administration improved spatial learning and memory of ETS mice (p < 0.05 or p < 0.01). Furthermore, exposure to ATX prevented the increases in the protein levels of the p38mitogen-activated protein kinase (p38 MAPK; p < 0.05 or p < 0.01) and nuclear factor-kappa B (NF-κB p65; p < 0.05 or p < 0.01), reversed the decreases in the mRNA and protein levels of synapsin I (SYN) and postsynaptic density protein 95 (PSD-95) (all p < 0.05 or p < 0.01). Moreover, ATX significantly down-regulated the increased levels of pro-inflammatory cytokines including interleukin-6 (IL-6) and tumor necrosis factor (TNF-α) (all p < 0.05 or p < 0.01). Meanwhile, the increased level of malondialdehyde (MDA) and the decreased activities of superoxide dismutase (SOD), glutathione (GSH), and catalase (CAT) were suppressed after exposure to ATX (all p < 0.05 or p < 0.01). Also, the results of the molecular docking study of ATX into the p38 MAPK binding site revealed that its mechanism was possibly similar to that of PH797804, a p38 MAPK inhibitor. Therefore, our results indicated that the ATX might be a critical agent in protecting the brain against neuroinflammation, synaptic plasticity impairment, and oxidative stress in the cortex and hippocampus of ETS mice.


Subject(s)
Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Nicotiana/adverse effects , Smoke/adverse effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Catalase/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cognition/drug effects , Cognitive Dysfunction/metabolism , Cytokines/metabolism , Glutathione/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Interleukin-6/metabolism , Male , Malondialdehyde/metabolism , Mice , NF-kappa B/metabolism , Oxidative Stress/drug effects , Tumor Necrosis Factor-alpha/metabolism , Xanthophylls/pharmacology
10.
Cancer Manag Res ; 10: 4969-4980, 2018.
Article in English | MEDLINE | ID: mdl-30464597

ABSTRACT

BACKGROUND: Glucose-6-phosphate isomerase (GPI) is a glycolytic-related enzyme that inter-converts glucose-6-phosphate and fructose-6-phosphate in the cytoplasm. This protein is also secreted into the extracellular matrix by cancer cells and is, therefore, also called autocrine motility factor (AMF). METHODS: To clarify the roles of AMF/GPI in gastric cancer (GC), we collected 335 GC tissues and the corresponding adjacent noncancerous tissues, performed immunohistochemical studies, and analyzed the relationship between AMF/GPI expression and the patients' clinicopathologic features. RESULTS: AMF/GPI expression was found to be significantly higher in the GC group than in the corresponding noncancerous tissue group (P<0.001). Additionally, AMF/GPI expression positively associated with a higher TNM stage and poorer prognosis in patients. Through Kaplan-Meier analysis and according to the Oncomine database, we found that AMF/GPI was overexpressed in GC tissues compared to normal mucosa, and the patients with higher AMF/GPI expression had poorer outcomes. We used AMF/GPI-silenced GC cell lines to observe how changes in AMP/GPI affect cellular phenotypes. AMF/GPI knockdown suppressed proliferation, migration, invasion, and glycolysis, and induced apoptosis in GC cells. CONCLUSION: These findings suggest that AMF/GPI overexpression is involved in carcinogenesis and promotes the aggressive phenotypes of GC cells.

11.
Neurochem Res ; 43(10): 1905-1913, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30088237

ABSTRACT

Luteolin, a flavonoid isolated from Cirsium japonicum, has antioxidant, anti-inflammatory and neuroprotective activities. Our previous studies brought a prospect that luteolin benefited diabetic rats with cognitive impairments. In this study, we examined whether luteolin could suppress the inflammatory cytokines, thus increasing synapse-associated proteins in streptozotocin (STZ)-induced diabetes in rat models. The model rats underwent luteolin treatment for 8 consecutive weeks, followed by assessment of cognitive performances with MWM test. Nissl staining was employed to assess the neuropathological changes in the hippocampus and the effects of luteolin on diabetic rats. With animals sacrificed, expressions of inflammatory cytokines including interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) and synapse-associated proteins including growth-associated protein-43 (GAP-43) and synaptophysin (SYN) were determined. The results affirmed improvement of behavioral performances in the MWM test, downexpression of glycation end products (AGEs) in the plasma and the receptor for advanced glycation end products in the hippocampus, inhibition of IL-1ß and TNF-α in both the hippocampus and plasma in diabetic rats. Furthermore, luteolin treatment upregulated the expressions of GAP-43 and SYN in the hippocampus. Thus, luteolin could ameliorate the cognitive dysfunctions in STZ-induced diabetic rat model.


Subject(s)
Cognitive Dysfunction/drug therapy , GAP-43 Protein/drug effects , Luteolin/pharmacology , Synaptophysin/metabolism , Animals , Cognitive Dysfunction/metabolism , Cytokines/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , GAP-43 Protein/metabolism , Inflammation/drug therapy , Male , Rats, Sprague-Dawley , Streptozocin/pharmacology , Synaptophysin/drug effects
12.
Gastric Cancer ; 21(1): 41-54, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28493075

ABSTRACT

BACKGROUND: We recently reported that miR-1 was one of the most significantly downregulated microRNAs in gastric cancer (GC) patients from The Cancer Genome Atlas microRNA sequencing data. Here we aim to elucidate the role of miR-1 in gastric carcinogenesis. METHODS: We measured miR-1 expression in human GC cell lines and 90 paired primary GC samples, and analyzed the association of its status with clinicopathological features. The effect of miR-1 on GC cells was evaluated by proliferation and migration assay. To identify the target genes of miR-1, bioinformatic analysis and protein array analysis were performed. Moreover, the regulation mechanism of miR-1 with regard to these predicted targets was investigated by quantitative PCR (qPCR), Western blot, ELISA, and endothelial cell tube formation. The putative binding site of miR-1 on target genes was assessed by a reporter assay. RESULTS: Expression of miR-1 was obviously decreased in GC cell lines and primary tissues. Patients with low miR-1 expression had significantly shorter overall survival compared with those with high miR-1 expression (P = 0.0027). Overexpression of miR-1 in GC cells inhibited proliferation, migration, and tube formation of endothelial cells by suppressing expression of vascular endothelial growth factor A (VEGF-A) and endothelin 1 (EDN1). Conversely, inhibition of miR-1 with use of antago-miR-1 caused an increase in expression of VEGF-A and EDN1 in nonmalignant GC cells or low-malignancy GC cells. CONCLUSIONS: MiR-1 acts as a tumor suppressor by inhibiting angiogenesis-related growth factors in human gastric cancer. Downregulated miR-1 not only promotes cellular proliferation and migration of GC cells, but may activates proangiogenesis signaling and stimulates the proliferation and migration of endothelial cells, indicating the possibility of new strategies for GC therapy.


Subject(s)
Adenocarcinoma/pathology , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Stomach Neoplasms/pathology , Adenocarcinoma/genetics , Adult , Aged , Endothelin-1/biosynthesis , Female , Genes, Tumor Suppressor , Humans , Male , Middle Aged , Stomach Neoplasms/genetics , Vascular Endothelial Growth Factor A/biosynthesis
13.
Oncol Lett ; 13(1): 222-230, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28123545

ABSTRACT

S100A6 is involved in regulating the progression of cancer. S100A6 can regulate the dynamics of cytoskeletal constituents, cell growth and differentiation by interacting with binding or target proteins. The present study investigated whether S100A6 affects cell proliferation in gastric cancer cells by stimulating several downstream factors. Firstly, the expression and localization of S100A6 were investigated using immunohistochemical staining, an immunoelectron microscopy and laser confocal scanning. A ChIP-Chip assay was performed to determine the downstream factors of S100A6 using promoter Chip analysis, including approximately the -800 to +200 regions around the transcription starting point. Polymerase chain reaction analysis was performed to confirm this. It was found that the intensity of S100A6 staining was markedly higher in the cytoplasm and nucleus, and its expression level correlated with that of the Ki67 protein. The overexpression of S100A6 also promoted cell proliferation in AGS and BGC823 cell lines, detected using a Cell Counting-Kit 8 assay. In cells overexpressing S100A6, the expression levels of interleukin (IL)-8, cyclin-dependent kinase (CDK)5, CDK4, minichromosome maintenance complex component 7 (MCM7) and B-cell lymphoma 2 (Bcl2) were noticeably increased. In conclusion, the increased expression of S100A6 promoted cell proliferation by regulating the expression levels of IL-8, CDK5, CDK4, MCM7 and Bcl2 in gastric cancer cells.

14.
Oncol Rep ; 35(5): 3009-17, 2016 May.
Article in English | MEDLINE | ID: mdl-26986870

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) holds promise for cancer therapy due to its unique capacity to selectively trigger apoptosis in cancer cells. However, TRAIL therapy is greatly hampered by its resistance. A preclinical successful strategy is to identify combination treatments that sensitize resistant cancers to TRAIL. In the present study, we fully assessed TRAIL sensitivity in 9 gastric cancer cell lines. We found combined administration of paclitaxel (PTX) markedly enhanced TRAIL-induced apoptosis in resistant cancer cells both in vitro and in vivo. The sensitization to TRAIL was accompanied by activation of mitochondrial apoptotic pathway, upregulation of TRAIL receptors and downregulation of anti-apoptotic proteins including C-IAP1, C-IAP2, Livin and Mcl-1. Noticeably, we found PTX could suppress the activation of mitogen-activated protein kinases (MAPKs). Inhibition of MAPKs using specific inhibitors (ERK inhibitor U0126, JNK inhibitor SP600125 and P38 inhibitor SB202190) facilitated TRAIL-mediated apoptosis and cytotoxicity. Additionally, SP600125 upregulated TRAL receptors as well as downregulated C-IAP2 and Mcl-1 suggesting the anti-apoptotic role of JNK. Thus, PTX-induced suppression of MAPKs may contribute to restoring TRAIL senstitivity. Collectively, our comprehensive analyses gave new insight into the role of PTX on enhancing TRAIL sensitivity, and provided theoretical references on the development of combination treatment in TRAIL-resistant gastric cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , MAP Kinase Signaling System/drug effects , Stomach Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Drug Synergism , Female , Humans , Mice, Nude , Paclitaxel/administration & dosage , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Tumour Biol ; 37(8): 10269-78, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26831669

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an ideal apoptosis inducer and believed to have promise in cancer therapy, yet part of cancer cells exhibit resistance to TRAIL-mediated apoptosis. This necessitates the exploration of agents that resensitizes cancer cells to TRAIL. In our study, we found that Trichostatin A (TSA), an histone deacetylase (HDAC) inhibitor, augmented TRAIL-induced apoptosis in gastric cancer cells in a caspase-dependent manner. Besides, upregulation of DR5 and downregulation of anti-apoptotic proteins including XIAP, Mcl-1, Bcl-2 and Survivin also contributed to this synergism. Noticeably, TSA treatment inhibited Forkhead boxM1 (FOXM1), which expression level showed negative correlation with TRAIL sensitivity. Similarly, silencing of FOXM1 by small interfering RNA (siRNA) resensitized cancer cells to TRAIL and strengthened the TRAIL-augmenting effect of TSA. In addition, we demonstrated the depletion of FOXM1 was a consequence of the inactivation of ERK mediated by TSA. Collectively, it was first shown that TSA potentiated TRAIL sensitivity via ERK/FOXM1 pathway in gastric cancer cells. FOXM1 might serve as a biomarker for predicting sensitivity to TRAIL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Hydroxamic Acids/pharmacology , Stomach Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Drug Synergism , Forkhead Box Protein M1/metabolism , Gene Knockdown Techniques , Histone Deacetylase Inhibitors/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism
16.
Am J Cancer Res ; 5(2): 589-602, 2015.
Article in English | MEDLINE | ID: mdl-25973299

ABSTRACT

BACKGROUND: The SUMO pathway has been shown to play an important role in tumorigenesis. This report analyzed the involvement of the sole SUMO-Activating Enzyme Subunit 2 (SAE2) in human gastric cancer (GC) progression and prognosis. METHODS: Expression of SAE2 was examined by Quantigene Plex, western blotting and immunohistochemistry. The expression of SAE2 and c-MYC were detected in parallel in 276 cases. The molecular mechanisms of SAE2 expression and its effects on cell growth, colony formation, migration and invasion were also explored by CCK8 assay, colony formation experiment, transwell chamber assay with or without matrigel, immunoprecipitation and in vivo tumorigenesis and tumor metastasis. RESULTS: SAE2 was markedly overexpressed in GC cell lines and primary tumor samples of GC, and significantly correlated with deeper tumor depth, distant metastasis, higher pathological stage and stratified survival in human GC. SAE2 positivity was independently associated with a worse outcome in multivariate analysis. Knockdown of SAE2 expression inhibited the proliferation, migration, and invasion of SAE2-overexpressing GC cells. Consistent with the in vitro results, down-regulation of SAE2 in human GC BGC823 cells significantly reduced the tumorigenic and metastatic potential of the cells in vivo. SAE2 protein was significantly associated with the higher expression of c-MYC in primary GC tissues. Moreover, FoxM1 was SUMOylated in GC and that inhibition of SAE2 resulted in a decrease in SUMO1-FoxM1 levels compared with those in the controls. CONCLUSIONS: These findings suggest that SAE2 has a pivotal role in the aggressiveness of GC, and highlight its usefulness as a prognostic factor in GC.

17.
Am J Cancer Res ; 5(1): 140-54, 2015.
Article in English | MEDLINE | ID: mdl-25628926

ABSTRACT

BACKGROUND: The SUMO pathway has been shown to play an important role in tumorigenesis. This report analyzed the involvement of the sole SUMO-Activating Enzyme Subunit 2 (SAE2) in human gastric cancer (GC) progression and prognosis. METHODS: Expression of SAE2 was examined by Quantigene Plex, western blotting and immunohistochemistry. The expression of SAE2 and c-MYC were detected in parallel in 276 cases. The molecular mechanisms of SAE2 expression and its effects on cell growth, colony formation, migration and invasion were also explored by CCK8 assay, colony formation experiment, transwell chamber assay with or without matrigel, immunoprecipitation and in vivo tumorigenesis and tumor metastasis. RESULTS: SAE2 was markedly overexpressed in GC cell lines and primary tumor samples of GC, and significantly correlated with deeper tumor depth, distant metastasis, higher pathological stage and stratified survival in human GC. SAE2 positivity was independently associated with a worse outcome in multivariate analysis. Knockdown of SAE2 expression inhibited the proliferation, migration, and invasion of SAE2-overexpressing GC cells. Consistent with the in vitro results, down-regulation of SAE2 in human GC BGC823 cells significantly reduced the tumorigenic and metastatic potential of the cells in vivo. SAE2 protein was significantly associated with the higher expression of c-MYC in primary GC tissues. Moreover, FoxM1 was SUMOylated in GC and that inhibition of SAE2 resulted in a decrease in SUMO1-FoxM1 levels compared with those in the controls. CONCLUSIONS: These findings suggest that SAE2 has a pivotal role in the aggressiveness of GC, and highlight its usefulness as a prognostic factor in GC.

18.
World J Gastroenterol ; 20(30): 10585-90, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25132779

ABSTRACT

AIM: To study the diagnostic value of controlled attenuation parameter (CAP), evaluated by transient elastography, for liver steatosis in patients with chronic hepatitis B (CHB). METHODS: Eighty-eight patients with CHB were enrolled in this study. All of the patients were subjected to transient elastography to determine CAP. These patients also underwent liver biopsy in the same period. Using liver biopsy as a reference, we determined receiver operating characteristic (ROC) curves for different endpoints. Areas under the ROC curves (AUCs) were used to evaluate the diagnostic importance of CAP for liver steatosis in patients with CHB. RESULTS: A positive correlation was observed between the AUCs of CAP and liver pathological stage (r = 0.582, P < 0.05). CAP was not correlated with inflammation degree and fibrosis degree (r = -0.025, P > 0.05; r = 0. 068, P > 0.05). The mean CAP value at S0 was 209.59 ± 41.25 dB/m, 223.84 ± 35.28 dB/m at S1, 274.17 ± 43.69 dB/m at S2, and 312.50 ± 25.44 dB/m at S3. CAP values among S0, S1, S2, and S3 were significantly different (F = 17.79, P < 0.01). The AUC values for CAP were 0.711 (0.592-0.870), 0.868 (0.748-0.989), and 0.974 (0.922-1.026) for S1, S2, and S3, respectively. The optimal cut-off values were 219.5, 230.0, and 283.5 dB/m. CONCLUSION: CAP is a novel tool that can be used to assess the degree of steatosis.


Subject(s)
Elasticity Imaging Techniques/methods , Fatty Liver/pathology , Hepatitis B, Chronic/complications , Liver/pathology , Adolescent , Adult , Aged , Area Under Curve , Biopsy , Elasticity , Fatty Liver/virology , Female , Hepatitis B, Chronic/diagnosis , Humans , Liver/virology , Liver Cirrhosis/pathology , Liver Cirrhosis/virology , Male , Middle Aged , Predictive Value of Tests , ROC Curve , Severity of Illness Index , Young Adult
19.
Zhonghua Wei Chang Wai Ke Za Zhi ; 16(2): 179-82, 2013 Feb.
Article in Chinese | MEDLINE | ID: mdl-23446483

ABSTRACT

OBJECTIVE: To explore the expression of CCAAT/enhancer binding protein beta (CEBPB) in gastric carcinoma tissues and its association with clinicopathological features and prognosis. METHODS: CEBPB protein expression level was detected by immunohistochemistry method in resected gastric carcinomas and adjacent gastric mucosa tissues (n=81), and its association with clinicopathological features and prognosis was analyzed. RESULTS: The immunohistochemical staining of CEBPB was predominantly in the nucleus with some cytoplasmic staining. As a result, 16% (13/81) of the gastric carcinomas were stained positively, whereas there was hardly positive expression in adjacent gastric mucosa tissues. There was a significant association between the expression of CEBPB and distant metastasis on univariate analysis (P<0.05). The median survival time in patients with positive CEBPB expression was significantly lower than those with negative CEBPB expression (19.4 months vs. 45.2 months, P=0.024). Multivariable analysis showed that CEBPB was independently associated with prognosis (HR=2.544, 95%CI:1.154-5.610, P=0.021). CONCLUSION: Up-regulation of CEBPB suggests poor prognosis in patients with gastric cancer.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Stomach Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Female , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Humans , Male , Middle Aged , Prognosis , Stomach Neoplasms/pathology
20.
Zhonghua Yi Xue Za Zhi ; 93(42): 3369-74, 2013 Nov 12.
Article in Chinese | MEDLINE | ID: mdl-24418034

ABSTRACT

OBJECTIVE: To explore the expressions of S100A8 and S100A9 in gastric cancer. METHODS: A total of 176 patients with gastric cancer, including 124 males and 52 females, were recruit from 1998 to 2004, average age 57(26-80)years. The expressions of S100A8 (n = 125) , S100A9 (n = 176) and S100A8/A9 heterodimer (calprotectin) in gastric tissue samples were assessed by immunohistochemistry and immunofluorescence. The co-localization of S100A9 and its dimerization partner S100A8 and heterodimer S100A8/A9 were examined by laser confocal scanning.Receiver operator characteristic (ROC) curve was used in determining the cut-off value of S100A9 and S100A8-positive inflammatory cell counts in evaluating the pathological TNM stage. To obtain associations between S100A9 or S100A8 cell counts and clinicopathologic variables, the data were cross-tabulated and χ(2)-test was performed. Cumulative survival was estimated by the Kaplan-Meier method. RESULTS: ROC curves using the S100A9 and S100A8-positive inflammatory cell counts were 0.623 and 0.522 for pathological TNM stages respectively. The cutoff values were 200 and 65 per 200× magnification field with a sensitivity of 61.48% and 51.09% and a specificity of 64.29% and 51.52% respectively. Patients with S100A9 positive expression (n = 77) had better overall survival than negative expression(n = 99) ((35.1 ± 10.8) vs (20.3 ± 3.0) months, P = 0.021). There was no statistical significance between S100A8 positive expression(n = 62) and negative expression(n = 63) ((26.4 ± 2.8) vs (29.5 ± 2.9) months, P = 0.145).In gastric cancer tissues, both S100A9 and S100A8 proteins were detected in tumor-infiltrating inflammatory cells while no case of S100A8/A9 heterodimer was found. In addition, S100A9 and S100A8 proteins were detected in inflammatory cells in chronic gastritis. Distribution of these two proteins also partly overlapped. CONCLUSIONS: S100A9 positive expression in gastric cancer tissues is associated with an excellent prognosis. But S100A8 positive expression has no prognostic association. Calprotectin expression differs between gastric cancer and gastritis.Further explorations are warranted.


Subject(s)
Calgranulin A/metabolism , Calgranulin B/metabolism , Stomach Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Female , Humans , Leukocyte L1 Antigen Complex/metabolism , Male , Middle Aged , Neoplasm Staging , Stomach Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...