Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cancer Commun (Lond) ; 41(3): 258-272, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33570246

RESUMO

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.


Assuntos
Insulinas , Neoplasias Gástricas , Carcinogênese , Linhagem Celular Tumoral , Proteínas Facilitadoras de Transporte de Glucose , Glicólise/genética , Humanos , Neoplasias Gástricas/genética
2.
J Cell Mol Med ; 24(21): 12355-12367, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32961025

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Curcumina/farmacologia , Cardiomiopatias Diabéticas/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Glicemia/metabolismo , Sobrevivência Celular , Diabetes Mellitus Experimental , Masculino , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Sirtuína 1/metabolismo
3.
Cancer Manag Res ; 11: 5375-5391, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31354346

RESUMO

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.

4.
ACS Chem Neurosci ; 10(8): 3500-3509, 2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31244052

RESUMO

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.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Inibidores da Colinesterase/uso terapêutico , Cognição/efeitos dos fármacos , Disfunção Cognitiva/tratamento farmacológico , Sulfeto de Hidrogênio/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Tacrina/uso terapêutico , Acetilcolinesterase/metabolismo , Cloreto de Alumínio , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/metabolismo , Animais , Inibidores da Colinesterase/farmacologia , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Combinação de Medicamentos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Sulfeto de Hidrogênio/farmacologia , Masculino , Memória/efeitos dos fármacos , Camundongos , Atividade Motora/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Tacrina/farmacologia
5.
Mar Drugs ; 17(1)2019 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-30609815

RESUMO

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.


Assuntos
Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Nicotiana/efeitos adversos , Fumaça/efeitos adversos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Catalase/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Cognição/efeitos dos fármacos , Disfunção Cognitiva/metabolismo , Citocinas/metabolismo , Glutationa/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Interleucina-6/metabolismo , Masculino , Malondialdeído/metabolismo , Camundongos , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Xantofilas/farmacologia
6.
Cell Death Dis ; 10(2): 33, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30674889

RESUMO

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.


Assuntos
Histona-Lisina N-Metiltransferase/genética , Proteínas com Homeodomínio LIM/biossíntese , Neoplasias Gástricas/genética , Fatores de Transcrição/biossíntese , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Animais , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Células HEK293 , Xenoenxertos , Humanos , Proteínas com Homeodomínio LIM/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Fatores de Transcrição/genética
7.
Cancer Manag Res ; 10: 4969-4980, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30464597

RESUMO

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.

8.
Neurochem Res ; 43(10): 1905-1913, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30088237

RESUMO

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.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Proteína GAP-43/efeitos dos fármacos , Luteolina/farmacologia , Sinaptofisina/metabolismo , Animais , Disfunção Cognitiva/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Proteína GAP-43/metabolismo , Inflamação/tratamento farmacológico , Masculino , Ratos Sprague-Dawley , Estreptozocina/farmacologia , Sinaptofisina/efeitos dos fármacos
9.
Gastric Cancer ; 21(1): 41-54, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28493075

RESUMO

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.


Assuntos
Adenocarcinoma/patologia , Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/genética , Neovascularização Patológica/genética , Neoplasias Gástricas/patologia , Adenocarcinoma/genética , Adulto , Idoso , Endotelina-1/biossíntese , Feminino , Genes Supressores de Tumor , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Gástricas/genética , Fator A de Crescimento do Endotélio Vascular/biossíntese
10.
Oncol Lett ; 13(1): 222-230, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28123545

RESUMO

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.

11.
Oncol Rep ; 35(5): 3009-17, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26986870

RESUMO

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.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias Gástricas/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Feminino , Humanos , Camundongos Nus , Paclitaxel/administração & dosagem , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Ligante Indutor de Apoptose Relacionado a TNF/administração & dosagem , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Tumour Biol ; 37(8): 10269-78, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26831669

RESUMO

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.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ácidos Hidroxâmicos/farmacologia , Neoplasias Gástricas/patologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Sinergismo Farmacológico , Proteína Forkhead Box M1/metabolismo , Técnicas de Silenciamento de Genes , Inibidores de Histona Desacetilases/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
13.
Am J Cancer Res ; 5(2): 589-602, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25973299

RESUMO

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.

14.
Am J Cancer Res ; 5(1): 140-54, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25628926

RESUMO

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.

15.
World J Gastroenterol ; 20(30): 10585-90, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25132779

RESUMO

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.


Assuntos
Técnicas de Imagem por Elasticidade/métodos , Fígado Gorduroso/patologia , Hepatite B Crônica/complicações , Fígado/patologia , Adolescente , Adulto , Idoso , Área Sob a Curva , Biópsia , Elasticidade , Fígado Gorduroso/virologia , Feminino , Hepatite B Crônica/diagnóstico , Humanos , Fígado/virologia , Cirrose Hepática/patologia , Cirrose Hepática/virologia , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Curva ROC , Índice de Gravidade de Doença , Adulto Jovem
16.
Zhonghua Wei Chang Wai Ke Za Zhi ; 16(2): 179-82, 2013 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-23446483

RESUMO

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.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Neoplasias Gástricas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Neoplasias Gástricas/patologia
17.
World J Gastroenterol ; 19(46): 8714-21, 2013 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-24379591

RESUMO

AIM: To investigate the differential expression of leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) in gastric cancer tissues and its significance related to tumor growth and spread. METHODS: Formalin-fixed biopsy specimens of intestinal metaplasia (n = 90), dysplasia (n = 53), gastric adenocarcinoma (n = 180), metastases in lymph nodes and the liver (n = 15), and lesion-adjacent normal gastric mucosa (controls; n = 145) were obtained for analysis from the Peking University Cancer Hospital's Department of Pathology and Gastrointestinal Surgery tissue archives (January 2003 to December 2011). The biopsied patients' demographic and clinicopathologic data were retrieved from the hospital's medical records database. Each specimen was subjected to histopathological typing to classify the tumor node metastasis (TNM) stage and to immunohistochemistry staining to detect the expression of the cancer stem cell marker LGR5. The intergroup differences in LGR5 expression were assessed by Spearman's rank correlation analysis, and the relationship between LGR5 expression level and the patients' clinicopathological characteristics was evaluated by the χ(2) test or Fisher's exact test. RESULTS: Significantly more gastric cancer tissues showed LGR5(+) staining than normal control tissues (all P < 0.01), with immunoreactivity detected in 72.2% (65/90) and 50.9% (27/53) of intestinal metaplasia and dysplasia specimens, respectively, 52.8% (95/180) of gastric adenocarcinoma specimens, and 73.3%% (11/15) of metastasis specimens, but 26.9% (39/145) of lesion-adjacent normal gastric mucosa specimens. Comparison of the intensity of LGR5(+) staining showed an increasing trend that generally followed increasing dedifferentiation and tumor spread (normal tissue < dysplasia, < gastric adenocarcinoma

Assuntos
Adenocarcinoma/química , Biomarcadores Tumorais/análise , Células-Tronco Neoplásicas/química , Receptores Acoplados a Proteínas G/análise , Neoplasias Gástricas/química , Adenocarcinoma/secundário , Adenocarcinoma/terapia , Biópsia , Desdiferenciação Celular , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/química , Neoplasias Hepáticas/secundário , Masculino , Metaplasia , Pessoa de Meia-Idade , Invasividade Neoplásica , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Células-Tronco Neoplásicas/patologia , Prognóstico , Neoplasias Gástricas/patologia , Neoplasias Gástricas/terapia , Regulação para Cima
18.
Zhonghua Yi Xue Za Zhi ; 93(42): 3369-74, 2013 Nov 12.
Artigo em Chinês | MEDLINE | ID: mdl-24418034

RESUMO

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.


Assuntos
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Neoplasias Gástricas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Complexo Antígeno L1 Leucocitário/metabolismo , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Gástricas/patologia
19.
BMC Cancer ; 12: 316, 2012 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-22838504

RESUMO

BACKGROUND: S100A9 was originally discovered as a factor secreted by inflammatory cells. Recently, S100A9 was found to be associated with several human malignancies. The purpose of this study is to investigate S100A9 expression in gastric cancer and explore its role in cancer progression. METHODS: S100A9 expression in gastric tissue samples from 177 gastric cancer patients was assessed by immunohistochemistry. The expression of its dimerization partner S100A8 and the S100A8/A9 heterodimer were also assessed by the same method. The effect of exogenous S100A9 on motility of gastric cancer cells AGS and BGC-823 was then investigated. RESULTS: S100A9 was specifically expressed by inflammatory cells such as macrophages and neutrophils in human gastric cancer and gastritis tissues. Statistical analysis showed that a high S100A9 cell count (> = 200) per 200x magnification microscopic field in cancer tissues was predictive of early stage gastric cancer. High S100A9-positive cell count was negatively correlated with lymph node metastasis (P = 0.009) and tumor invasion (P = 0.011). S100A9 was identified as an independent prognostic predictor of overall survival of patients with gastric cancer (P = 0.04). Patients with high S100A9 cell count were with favorable prognosis (P = 0.021). Further investigation found that S100A8 distribution in human gastric cancer tissues was similar to S100A9. However, the number of S100A8-positive cells did not positively correlate with patient survival. The inflammatory cells infiltrating cancer were S100A8/A9 negative, while those in gastritis were positive. Furthermore, exogenous S100A9 protein inhibited migration and invasion of gastric cancer cells. CONCLUSIONS: Our results suggested S100A9-positive inflammatory cells in gastric cancer tissues are associated with early stage of gastric cancer and good prognosis.


Assuntos
Calgranulina B/imunologia , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Calgranulina A/imunologia , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Feminino , Humanos , Inflamação/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neutrófilos/imunologia , Neutrófilos/metabolismo , Prognóstico , Multimerização Proteica , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidade
20.
J Surg Oncol ; 106(7): 880-6, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22585737

RESUMO

BACKGROUND AND OBJECTIVES: Protein tyrosine kinase 7 (PTK7) plays important functions in several cancer types but its expression in gastric cancer remains unknown. This study was designed to investigate PTK7 expression in gastric cancer. METHODS: PTK7 expression was assessed by immunohistochemistry in 201 gastric cancer patients. The relationship between PTK7 expression and clinicopathological features and patients prognosis were statistically analyzed. RESULTS: PTK7 expression was detected in 56.72% (114 of 201) of gastric cancer patients. The immunostaining was predominantly localized in the cytoplasm. The statistical analyses showed that PTK7 expression was more frequently detected in patients with well-differentiated tumors (P = 0.001). Furthermore, PTK7 expression was significantly related to the favorable overall survival (OS; P = 0.012) and disease-free survival (DFS; P = 0.009). Multivariate Cox regression analyses revealed that PTK7 expression was an independent prognostic factor for both favorable OS (P = 0.028) and DFS (P = 0.012). CONCLUSION: Our findings demonstrate that PTK7 can serve as a novel prognostic biomarker for gastric cancer patients.


Assuntos
Biomarcadores Tumorais/metabolismo , Moléculas de Adesão Celular/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidade , Idoso , Estudos de Casos e Controles , Estudos de Coortes , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Prognóstico , Neoplasias Gástricas/patologia , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA