Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1393801, 2024.
Article in English | MEDLINE | ID: mdl-38660302

ABSTRACT

Background: Human tumors pose significant challenges, with targeted therapy against specific molecular targets or signaling pathways being a mainstay alongside surgical resection. Previous studies have implicated KHDRBS1 in the oncogenesis of certain human tumors such as colorectal and prostate cancers, underscoring its potential as a therapeutic target. However, the comprehensive expression pattern of KHDRBS1 in hepatocellular carcinoma (HCC) warrants further exploration. Methods: Integrating and analyzing multi-omics, multi-cohort data from public databases, coupled with clinical samples and molecular biology validation, we elucidate the oncogenic role of KHDRBS1 in HCC progression. Additionally, leveraging HCC single-cell sequencing data, we segregate malignant cells into KHDRBS1-positive and negative subsets, uncovering significant differences in their expression profiles and functional roles. Results: Our study identifies KHDRBS1 as a tumor-promoting factor in HCC, with its positivity correlating with tumor progression. Furthermore, we highlight the clinical significance of KHDRBS1-positive malignant cells, aiming to further propel its clinical utility. Conclusion: KHDRBS1 plays a key role in HCC development. This study provides crucial insights for further investigation into KHDRBS1 as a therapeutic target in HCC.


Subject(s)
Adaptor Proteins, Signal Transducing , Biomarkers, Tumor , Carcinoma, Hepatocellular , Liver Neoplasms , Tumor Microenvironment , Humans , Male , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/immunology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Prognosis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , Tumor Microenvironment/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
2.
Immunopharmacol Immunotoxicol ; 46(1): 117-127, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38047472

ABSTRACT

BACKGROUND: Splenomegaly can exacerbate liver cirrhosis and portal hypertension. We have previously demonstrated that cyclooxygenase-2 (COX-2) inhibitor can attenuate cirrhotic splenomegaly. However, the mechanism of cirrhotic splenomegaly remains unclear, thus becoming the focus of the present study. MATERIALS AND METHODS: Thioacetamide (TAA) intraperitoneal injection was used to induce cirrhotic splenomegaly. Rats were randomized into the control, TAA and TAA + celecoxib groups. Histological analysis and high-throughput RNA sequencing of the spleen were conducted. Splenic collagen III, α-SMA, Ki-67, and VEGF were quantified. RESULTS: A total of 1461 differentially expressed genes (DEGs) were identified in the spleens of the TAA group compared to the control group. The immune response and immune cell activation might be the major signaling pathways involved in the pathogenesis of cirrhotic splenomegaly. With its immunoregulatory effect, celecoxib presents to ameliorate cirrhotic splenomegaly and liver cirrhosis. Furthermore, 304 coexisting DEGs were obtained between TAA vs. control and TAA + celecoxib vs. TAA. Gene ontology (GO) and KEGG analyses collectively indicated that celecoxib may attenuate cirrhotic splenomegaly through the suppression of splenic immune cell proliferation, inflammation, immune regulation, and fibrogenesis. The impacts on these factors were subsequently validated by the decreased splenic Ki-67-positive cells, macrophages, fibrotic areas, and mRNA levels of collagen III and α-SMA. CONCLUSIONS: Celecoxib attenuates cirrhotic splenomegaly by inhibiting splenic immune cell proliferation, inflammation, and fibrogenesis. The current study sheds light on the therapeutic strategy of liver cirrhosis by targeting splenic abnormalities and provides COX-2 inhibitors as a novel medical treatment for cirrhotic splenomegaly.


Subject(s)
Liver Cirrhosis , Splenomegaly , Rats , Animals , Celecoxib/pharmacology , Splenomegaly/drug therapy , Splenomegaly/etiology , Splenomegaly/pathology , Ki-67 Antigen , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Collagen , Inflammation/drug therapy , Gene Expression Profiling
4.
Arch Toxicol ; 96(12): 3143-3162, 2022 12.
Article in English | MEDLINE | ID: mdl-36116095

ABSTRACT

The p21-activated kinase 1 (PAK1), an effector protein of the small G protein Rac and cell division cycle protein 42 (Cdc42), is highly expressed in cardiac tissue. Although a large number of studies have explored the molecular basis and biological function of PAK1, research on PAK1 as a therapeutic target for cardiotoxicity remains in a stage of continuous innovation, and further clarification of its role in cardiotoxicity is required. In this review, we examine the important role of PAK1 in the programmed death (apoptosis, autophagy, and pyroptosis) of cardiomyocytes, and its involvement in oxidative stress and inflammatory responses, which are based on mitochondrial dysfunction and calcium homeostasis imbalance. We also summarize the related signaling pathways through which PAK1 may cause oxidative stress and inflammatory response in cardiotoxicity, and discuss the PAK1-mediated contributions of the gut microbiome and micro RNAs to cardiotoxicity. We propose that PAK1 holds great promise for novel therapeutic strategies to facilitate improvements in the treatment of complex and diverse cardiovascular diseases.


Subject(s)
Cardiotoxicity , p21-Activated Kinases , Humans , p21-Activated Kinases/metabolism , Calcium , Myocytes, Cardiac/metabolism , Cell Cycle Proteins
5.
J Cell Mol Med ; 25(22): 10389-10402, 2021 11.
Article in English | MEDLINE | ID: mdl-34609050

ABSTRACT

The balance between endothelial nitric oxide (NO) synthase (eNOS) activation and production of reactive oxygen species (ROS) is very important for NO homeostasis in liver sinusoidal endothelial cells (LSECs). Overexpression of cyclooxygenase-2 (COX-2), a major intravascular source of ROS production, has been observed in LSECs of cirrhotic liver. However, the links between low NO bioavailability and COX-2 overexpression in LSECs are unknown. This study has confirmed the link between low NO bioavailability and COX-2 overexpression by COX-2-dependent PGE2-EP2-ERK1/2-NOX1/NOX4 signalling pathway in LSECs in vivo and in vitro. In addition, the regulation of COX-2-independent LKB1-AMPK-NRF2-HO-1 signalling pathway on NO homeostasis in LSECs was also elucidated. The combinative effects of celecoxib on diminishment of ROS via COX-2-dependent and COX-2-independent signalling pathways greatly decreased NO scavenging. As a result, LSECs capillarisation was reduced, and endothelial dysfunction was corrected. Furthermore, portal hypertension of cirrhotic liver was ameliorated with substantial decreasing hepatic vascular resistance and great increase of portal blood flow. With the advance understanding of the mechanisms of LSECs protection, celecoxib may serve as a potential therapeutic candidate for patients with cirrhotic portal hypertension.


Subject(s)
Celecoxib/therapeutic use , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Hypertension, Portal/drug therapy , Hypertension, Portal/metabolism , Oxidative Stress/drug effects , Vascular Resistance/drug effects , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Celecoxib/pharmacology , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Disease Management , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Heme Oxygenase (Decyclizing)/metabolism , Hemodynamics/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Hypertension, Portal/diagnosis , Hypertension, Portal/etiology , Male , Models, Biological , Nitric Oxide/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
6.
Exp Ther Med ; 22(5): 1213, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34584558

ABSTRACT

Chronic inflammation is considered as an important pathophysiologic mechanism of hepatic cirrhosis, which induces hepatocyte injury and activates hepatic stellate cells (HSCs), thus resulting in hepatic fibrosis. Previous studies have reported that cyclooxygenase-2 (COX-2) inhibitor can effectively treat liver fibrosis, while somatostatin (SST) analogues inhibit the activation of HSCs. The present study aimed to investigate the effects of a COX-2 inhibitor, celecoxib, combined with a SST analogue, octreotide, for protection of hepatocytes and prevention of fibrosis in a rat model of hepatic fibrosis. Therefore, a hepatic fibrosis rat model was established following peritoneal injection of thioacetamide (TAA), and the rats were then treated with a combination of celecoxib and octreotide (TAA + C). Immunohistochemistry and western blotting assays were used to assess the expression levels of proteins associated with inflammation, epithelial-mesenchymal transition (EMT), proliferation, apoptosis and autophagy. H&E staining, transmission electron microscopy and scanning electron microscopy were used to evaluate the destruction of hepatocytes. Masson's Trichrome and Sirius Red were used to measure the degree of liver fibrosis. The results demonstrated that, compared with those of the control group, the degree of liver fibrosis and the expression of the intrahepatic inflammation factors were aggravated in the TAA group. Furthermore, the apoptosis rate, EMT and autophagy of hepatocytes were also increased in the TAA group. However, treatment with TAA + C restored the aforementioned increased levels compared with the TAA group. In conclusion, treatment of rats with the combination of celecoxib and octreotide could attenuate the progress of hepatic fibrosis via protection of hepatocytes by reducing apoptosis, EMT and autophagy in hepatocytes.

7.
J Cell Mol Med ; 25(16): 7993-8005, 2021 08.
Article in English | MEDLINE | ID: mdl-34145945

ABSTRACT

The intestinal barrier dysfunction is crucial for the development of liver fibrosis but can be disturbed by intestinal chronic inflammation characterized with cyclooxygenase-2 (COX-2) expression. This study focused on the unknown mechanism by which COX-2 regulates intestinal epithelial homeostasis in liver fibrosis. The animal models of liver fibrosis induced with TAA were established in rats and in intestinal epithelial-specific COX-2 knockout mice. The impacts of COX-2 on intestinal epithelial homeostasis via suppressing ß-catenin signalling pathway were verified pharmacologically and genetically in vivo. A similar assumption was tested in Ls174T cells with goblet cell phenotype in vitro. Firstly, disruption of intestinal epithelial homeostasis in cirrhotic rats was ameliorated by celecoxib, a selective COX-2 inhibitor. Then, ß-catenin signalling pathway in cirrhotic rats was associated with the activation of COX-2. Furthermore, intestinal epithelial-specific COX-2 knockout could suppress ß-catenin signalling pathway and restore the disruption of ileal epithelial homeostasis in cirrhotic mice. Moreover, the effect of COX-2/PGE2 was dependent on the ß-catenin signalling pathway in Ls174T cells. Therefore, inhibition of COX-2 may enhance intestinal epithelial homeostasis via suppression of the ß-catenin signalling pathway in liver fibrosis.


Subject(s)
Celecoxib/pharmacology , Cyclooxygenase 2/chemistry , Homeostasis , Inflammation/drug therapy , Intestinal Mucosa/drug effects , Liver Cirrhosis/drug therapy , beta Catenin/metabolism , Animals , Cell Line , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Disease Models, Animal , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley , Signal Transduction , beta Catenin/genetics
8.
Life Sci ; 272: 119203, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33577848

ABSTRACT

BACKGROUND & AIMS: Splenomegaly is usually taken as a consequence of liver cirrhosis. However, as a risk factor for cirrhosis, the impacts of spleen-liver axis on the development of cirrhosis are largely unknown. This study focused on the impacts of splenomegaly on the development of cirrhosis and assessment of the effects of celecoxib, a selective COX-2 inhibitor, on the splenomegaly and cirrhotic liver. MATERIALS AND METHODS: Liver cirrhosis was induced by thioacetamide (TAA). Sixty rats were randomly divided into control, TAA-16w, TAA + celecoxib groups and normal, TAA + sham, TAA + splenectomy groups. Hepatic stellate cells (HSCs) or hepatocytes were co-cultured with splenocytes from those groups. RESULTS: Splenocytes of cirrhotic rats stimulated the HSCs activation and induced hepatocyte apoptosis via enhancing oxidative stress. The hepatic levels of NOX-4 and the in situ O2- were profoundly reduced in TAA + splenectomy group by 50.6% and 18.5% respectively, p < 0.05. Celecoxib significantly decreased the hepatic fibrotic septa induced with TAA by 50.8%, p < 0.05. Splenic lymphoid tissue proliferation and proinflammatory cytokines of the cirrhotic rats were also obviously suppressed by celecoxib, p < 0.05. Compared with the HSC or hepatocyte cell line co-cultured with the cirrhotic splenocytes, the expression of alpha-SMA, NOX-4, in situ O2- or the levels of cleaved caspase3 and NOX-4 were significantly decreased in those cell lines co-cultured with cirrhotic splenocytes treated by celecoxib, p < 0.05. CONCLUSION: Splenomegaly contributed to the development of liver cirrhosis through enhancing oxidative stress in liver. Celecoxib could effectively ameliorate liver cirrhosis via reducing inflammatory cytokines and immune cells derived from spleen and suppressing oxidative stress.


Subject(s)
Celecoxib/pharmacology , Liver Cirrhosis/drug therapy , Animals , Apoptosis/drug effects , Celecoxib/metabolism , China , Cyclooxygenase 2 Inhibitors/pharmacology , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Inflammation/drug therapy , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis, Experimental/metabolism , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spleen/pathology , Splenomegaly/complications , Splenomegaly/physiopathology , Thioacetamide/pharmacology
9.
Dig Dis Sci ; 66(1): 131-142, 2021 01.
Article in English | MEDLINE | ID: mdl-32144600

ABSTRACT

BACKGROUND: In liver cirrhosis, intestinal mucus barrier is rarely studied. AIMS: This study aimed to investigate whether mucus barrier in ileum is altered in cirrhotic rats and its underlying mechanisms. METHODS: Thioacetamide was injected to induce liver cirrhosis in rats. Serum from portal vein blood, and ileum and liver tissues were obtained for further analysis. Goblet cell-like Ls174T cells were cultured for in vitro experiments. RESULTS: The ileal mucus was thin, loose, and porous with small bubbles in cirrhotic rats. mRNA expressions of Muc2 and TFF3 were also down-regulated in cirrhotic rats. Bacteria located near to crypts and LPS were increased in the serum from portal vein in cirrhotic rats. Smaller theca area and few goblet cells were found in cirrhotic rats compared with control. Increased proliferation of ileal epithelia was observed in cirrhotic rats. Notch1, Dll1, and Hes1 expressions were enhanced, and KLF4 expression was suppressed in ileum of cirrhotic rats. In Ls174T cells, EDTA and NICD plasmid induced NICD and Hes1 expression and suppressed KLF4 concomitantly, and mucus expression almost vanished in these cells. NICD plasmid induced more proliferation in Ls174T cells. Oppositely, after DBZ treatment, NICD and Hes1 were inhibited along with augmentation of KLF4 and increased mucous expression in Ls174T cells, while proliferation of the cells was suppressed. CONCLUSIONS: In cirrhotic rats, mucus barrier was impaired. This might be attributed to increased proliferation and decreased differentiation of epithelia, which might be mediated by Notch1-Hes1-KLF4 signaling.


Subject(s)
Homeostasis/physiology , Ileum/metabolism , Intestinal Mucosa/metabolism , Liver Cirrhosis/metabolism , Receptor, Notch1/biosynthesis , Animals , Cell Line, Tumor , Homeostasis/drug effects , Humans , Ileum/drug effects , Ileum/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Kruppel-Like Factor 4 , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Rats , Rats, Sprague-Dawley , Thioacetamide/toxicity
10.
Int J Clin Exp Pathol ; 13(9): 2392-2396, 2020.
Article in English | MEDLINE | ID: mdl-33042349

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is a rare fatal hyperinflammatory syndrome resulting in cytokine storm and secondary multi-organ impairment. The natural killer (NK)/T-cell lymphoma is the predominant subtype in patients with lymphoma-associated hemophagocytic syndrome (LAHS) in Asia. Yet the non-Hodgkin's B-cell lymphoma is a relatively uncommon trigger of HLH. We report a case of a 64-year-old woman who had a bone marrow-spleen type of diffuse large B-cell lymphoma (DLBCL) associated with HLH. The patient presented with EBV-positive infection, significantly increased inflammatory cytokines (IL-6, IL-8, IL-10), and dramatically increased aspartate aminotransferase (AST) and total bilirubin (TB), resulting the patient's aggressive clinical course and early death. This case may not only illustrate the nonspecific manifestation and rapidly progressive characteristics of HLH but also highlight the necessity of anti-inflammatory therapy for the treatment of lymphoma-associated HLH.

11.
World J Gastroenterol ; 26(28): 4094-4107, 2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32821072

ABSTRACT

BACKGROUND: Endoplasmic reticulum (ER) stress is an important mechanism in the progression of chronic and acute liver diseases, especially in the progression and recovery of liver fibrosis. Excessive and long-term ER stress induces apoptosis. ER stress-induced apoptosis is considered to be an important pathway in the development of liver fibrosis. Cyclooxygenase-2 (COX-2) induction is also closely related to ER stress. In our previous studies, we showed that celecoxib, a COX-2 inhibitor, improves liver fibrosis and portal hypertension. However, the role and mechanism of celecoxib in alleviating liver fibrosis remain unclear. AIM: To investigate whether celecoxib alleviates liver fibrosis by inhibiting hepatocyte apoptosis via the ER stress response. METHODS: Cirrhosis was induced by intraperitoneal injections of thioacetamide (TAA) for 16 wk (injection dose is 200 mg/kg per 3 d for the first 8 wk and 100 mg /kg per 3 d after 8 wk). Thirty-six male Sprague-Dawley rats were randomly divided into three groups, namely, control group, TAA group, and TAA + celecoxib group. In the last 8 wk, TAA-induced cirrhotic rats received celecoxib (20 mg/kg/day) or the vehicle by gastric gavage. After 16 wk, the rats were sacrificed, and serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), and albumin (ALB) were detected. The hepatic fibrosis areas were evaluated by Sirius red staining and the degree of fibrosis was assessed by measuring the level of hydroxyproline. ER stress levels were evaluated by detecting the marker proteins glucose-regulated protein 78 (GRP78), CCAAT/enhancer binding protein homologous protein (CHOP), PKR-like ER protein kinase (PERK), activating transcription factor 6 (ATF6), and inositol-requiring enzyme 1 alpha (IRE1α). Apoptosis levels were evaluated by detecting caspase-12 and caspase-3. RESULTS: The serum ALT and AST levels in the liver were significantly reduced by celecoxib; however, the serum ALB had no significant changes. Celecoxib significantly reduced the degree of liver fibrosis and the levels of hydroxyproline (-38% and -25.7%, respectively, P < 0.01). Celecoxib ameliorated ER stress by reducing the level of GRP78 compared to the TAA group (P < 0.05). Consistently, after celecoxib administration, the upregulation of TAA-induced hepatic apoptosis markers (caspase-12 and caspase-3) and CHOP were significantly inhibited. In addition, after celecoxib treatment, the expression of key molecules associated with ER stress (PERK, ATF6, and IRE1) was decreased (P < 0.05). CONCLUSION: Therapeutic administration of celecoxib effectively reduces hepatic apoptosis in TAA-induced cirrhotic rats. The mechanism of action may be attributed to the suppression of CHOP expression, which subsequently inhibits ER stress.


Subject(s)
Endoplasmic Reticulum Stress , Thioacetamide , Animals , Apoptosis , Celecoxib/pharmacology , Endoribonucleases , Hepatocytes/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Male , Protein Serine-Threonine Kinases , Rats , Rats, Sprague-Dawley , Thioacetamide/toxicity
12.
Mol Med Rep ; 15(4): 1507-1512, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28259935

ABSTRACT

Multiple long non-coding RNAs (lncRNAs) have been demonstrated to be involved in liver disease. Increased cyclooxygenase-2 (COX­2) levels have also been reported to be involved in the progression of liver cirrhosis. In the present study, the correlations between lncRNA­COX­2 RNA expression levels, COX­2 mRNA expression levels and liver fibrosis were examined. Liver fibrosis was induced by intraperitoneal injection of carbon tetrachloride (CCl4) in mice for 2 months (CCl4­2M) or 3 months (CCl4­3M). Liver histopathological evaluation was conducted using hematoxylin and eosin and Masson trichrome staining. Hepatic expression of COX­2 and lncRNA­COX­2 was evaluated by reverse transcription­quantitative polymerase chain reaction and immunohistochemical staining. Compared with the control group, fibrotic areas were increased four and nine times in the CCl4­2M group and the CCl4­3M group, respectively. LncRNA-COX-2 and COX­2 upregulation were observed in the cirrhotic liver. COX­2 mRNA expression levels and lncRNA-COX-2 RNA expression levels were significantly positively correlated with the fibrotic area. In addition, COX­2 mRNA expression was significantly positively correlated with lncRNA­COX­2 expression. These results suggest that expression of COX­2 and lncRNA­COX­2 increased with the progression of liver fibrosis. LncRNA-COX-2 may potentially be considered as a novel therapeutic target for liver fibrosis.


Subject(s)
Cyclooxygenase 2/metabolism , Liver Cirrhosis/enzymology , Liver Cirrhosis/genetics , RNA, Long Noncoding/metabolism , Animals , Carbon Tetrachloride , Cyclooxygenase 2/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Mice, Inbred BALB C , RNA, Long Noncoding/genetics , Up-Regulation/genetics
13.
Int J Clin Exp Pathol ; 10(8): 8461-8469, 2017.
Article in English | MEDLINE | ID: mdl-31966698

ABSTRACT

Angiogenesis induced by vascular endothelial growth factor A (VEGF-A) plays a critical role in tumor growth and metastasis. The study aimed to evaluate the expression of VEGF-A in gastric adenocarcinoma and investigate its correlations with tumor clinicopathological features and prognostic significance. VEGF-A expression was detected by immunohistochemistry on a tissue microarray containing 90 pairs of human gastric adenocarcinoma and paracancerous tissues. Levels of VEGF-A in gastric adenocarcinoma were significantly higher than those in paracancerous tissues (P=0.018). Furthermore, the result was coincident with that of human gastric adenocarcinoma xenografts in nude-mice (P<0.01). In addition, the VEGF-A expression was positive correlation with TNM stage (P=0.047), tumor size (P=0.028), positive lymph nodes (P=0.002) and lymphovascular invasion (P=0.001). Finally, Kaplan-Meier survival analysis showed that VEGF-A up-regulation indicated a poor prognosis for overall survival (P=0.039). In conclusions, VEGF-A may be used as a biomarker for evaluating both the biological behavior of tumor and the prognosis in patients with gastric adenocarcinoma.

14.
Sci Rep ; 6: 35426, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27739503

ABSTRACT

Among the researches on hepatic fibrosis, great attention was paid to both hepatocytes and extracellular matrix (ECM). However, little focus was drawn on reticular fibrous network, which is important for demarcation and support of hepatocytes. The aim of this study was to investigate the change pattern of reticular fibers in hepatic fibrosis/cirrhosis and its underlying mechanism. In this study, thioacetamide (TAA) and bile duct ligation (BDL) were utilized to induce rat hepatic fibrosis respectively, and Human liver cirrhotic microassay was analyzed with IHC to confirm the results in animal experiment and to detect the metalloproteinases (MMPs) expressions. As a result, the reticular fibers decreased markedly after 1 week in TAA and 1 day in BDL treated rats. Multiple representative regulators of MMPs and MMPs increased significantly in their expressions and activities. Further more, in human liver cirrhotic microassay, MMPs expressions also showed similar patterns as that of animal experiment. In Conclusions: Degradation or collapse of reticular fibers in hepatic sinusoid can be considered as a pathological feature during the initiation and/or progression of hepatic fibrosis. Moreover, such degradation is associated with and probably caused by the over/dysregulated expression of MMPs.


Subject(s)
Extracellular Matrix/metabolism , Liver Cirrhosis/metabolism , Animals , Extracellular Matrix/ultrastructure , Humans , Liver/metabolism , Liver/ultrastructure , Liver Cirrhosis/pathology , Male , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Rats , Rats, Sprague-Dawley
15.
Angiogenesis ; 19(4): 501-11, 2016 10.
Article in English | MEDLINE | ID: mdl-27380212

ABSTRACT

Abnormal angiogenesis is critical for portal hypertension in cirrhosis. Except for etiological treatment, no efficient medication or regime has been explored to treat the early stage of cirrhosis when angiogenesis is initiated or overwhelming. In this study, we explored an anti-angiogenesis effort through non-cytotoxic drugs octreotide and celecoxib to treat early stage of cirrhotic portal hypertension in an animal model. Peritoneal injection of thioacetamide (TAA) was employed to induce liver cirrhosis in rats. A combination treatment of celecoxib and octreotide was found to relieve liver fibrosis, portal venous pressure, micro-hepatic arterioportal fistulas, intrahepatic and splanchnic angiogenesis. Celecoxib and octreotide exerted their anti-angiogenesis effect via an axis of cyclooxygenase-2/prostaglandin E2/EP-2/somatostatin receptor-2, which consequently down-regulated phosphorylation of extracellular signal-regulated kinase (p-ERK)-hypoxia-inducible factor-1α (HIF-1α)-vascular endothelial growth factor (VEGF) integrated signaling pathways. In conclusions, combination of celecoxib and octreotide synergistically ameliorated liver fibrosis and portal hypertension of the cirrhotic rats induced by TAA via the inhibition of intrahepatic and extrahepatic angiogenesis. The potential mechanisms behind the regimen may due to the inactivation of p-ERK-HIF-1α-VEGF signaling pathway.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Celecoxib/administration & dosage , Hypertension, Portal/prevention & control , Liver Cirrhosis, Experimental/complications , Liver Cirrhosis, Experimental/drug therapy , Neovascularization, Pathologic/prevention & control , Octreotide/administration & dosage , Animals , Cyclooxygenase 2 Inhibitors/administration & dosage , Drug Synergism , Hypertension, Portal/pathology , Hypertension, Portal/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver/blood supply , Liver/drug effects , Liver/pathology , Liver Cirrhosis, Experimental/pathology , MAP Kinase Signaling System/drug effects , Neovascularization, Pathologic/pathology , Portal Pressure/drug effects , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Thioacetamide/toxicity , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G962-72, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27056726

ABSTRACT

Inflammatory transport through the gut-liver axis may facilitate liver cirrhosis. Cyclooxygenase-2 (COX-2) has been considered as one of the important molecules that regulates intestinal epithelial barrier function. This study was aimed to test the hypothesis that inhibition of COX-2 by celecoxib might alleviate liver cirrhosis via reduction of intestinal inflammatory transport in thiacetamide (TAA) rat model. COX-2/prostaglandin E2 (PGE2)/EP-2/p-ERK integrated signal pathways regulated the expressions of intestinal zonula occludens-1 (ZO-1) and E-cadherin, which maintain the function of intestinal epithelial barrier. Celecoxib not only decreased the intestinal permeability to a 4-kDa FITC-dextran but also significantly increased expressions of ZO-1 and E-cadherin. When celecoxib greatly decreased intestinal levels of LPS, TNF-α, and IL-6, it significantly enhanced T cell subsets reduced by TAA. As a result, liver fibrosis induced by TAA was significantly alleviated in the celecoxib group. These data indicated that celecoxib improved the integrity of intestinal epithelial barrier, blocked inflammatory transport through the dysfunctional gut-liver axis, and ameliorated the progress of liver cirrhosis.


Subject(s)
Celecoxib/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Jejunum/metabolism , Liver Cirrhosis/drug therapy , Liver/metabolism , Animals , Caco-2 Cells , Cadherins/metabolism , Celecoxib/therapeutic use , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/therapeutic use , Dinoprostone/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interleukin-6/metabolism , Intestinal Absorption , Jejunum/drug effects , Liver/drug effects , Liver Cirrhosis/metabolism , Rats , Rats, Sprague-Dawley , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism , Zonula Occludens-1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...