Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Oncol Rep ; 52(3)2024 Sep.
Article in English | MEDLINE | ID: mdl-39027998

ABSTRACT

Following the publication of this article, an interested reader drew to the authors' attention that two pairs of protein bands featured in the western blots in Fig. 3A and 5D on p. 679 and 681 respectively appeared to be strikingly similar. After having re­examined their original data, the authors realized that Fig. 5D had been assembled incorrectly. The revised version of Fig. 5, now including the correct data for Fig. 5D, is shown on the next page. Note that the errors made in terms of assembling the data in Fig. 5 did not greatly affect either the results or the conclusions reported in this paper, and all the authors agree to the publication of this corrigendum. The authors regret that these errors went unnoticed prior to the publication of their article, are grateful to the Editor of Oncology Reports for allowing them this opportunity to publish this corrigendum. They also apologize to the readership for any inconvenience caused. [Oncology Reports  33: 675­684, 2015; DOI: 10.3892/or.2014.3653].

2.
BMC Cancer ; 24(1): 226, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38365617

ABSTRACT

BACKGROUND: Systemic inflammatory factors can predict the survival prognosis of gastric cancer (GC) patients after neoadjuvant chemotherapy (NACT). However, whether longitudinal changes in systemic inflammatory factors are associated with short - and long-term outcomes has not been reported. METHODS: This study is a retrospective analysis of 216 patients with advanced gastric cancer who received NACT between January 2011 and June 2019, comparing receiver operating characteristic (ROC) curves for screening suitable inflammatory markers. Group-based trajectory modeling (GBTM) was used to analyze longitudinal changes in inflammatory markers during NACT to identify different potential subgroups and to compare postoperative complications, recurrence-free survival (RFS), and overall survival (OS) among subgroups. RESULTS: Ultimately, neutrophil-lymphocyte ratio (NLR) had the highest area under the curve (AUC) value in predicting prognosis was included in the GBTM analysis. Three trajectories of NLR were obtained: Stable group (SG) (n = 89), Ascent-descend group (ADG) (n = 80) and Continuous descend group (CDG) (n = 47). Compared with SG, ADG and CDG are associated with an increased risk of postoperative recurrence and death. The median time of RFS and OS of SG was longer than that of ADG and CDG (median RFS 81 vs. 44 and 22 months; median OS 69 vs. 41 and 30 months). In addition, CDG had significantly higher postoperative serious complications than SG and ADG (17 (36.2%) vs. 17 (19.1%) and 12 (15.0%); p = 0.005). CONCLUSION: There were different trajectories of NLR during NACT, and these potential trajectories were significantly associated with severe postoperative complications, recurrence, and mortality in patients with GC.


Subject(s)
Neutrophils , Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/surgery , Retrospective Studies , Neoadjuvant Therapy , Lymphocytes , Prognosis , Postoperative Complications
3.
Ann Surg Oncol ; 31(4): 2679-2688, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38142258

ABSTRACT

BACKGROUND: Robotic gastrectomy (RG) has been widely used to treat gastric cancer. However, whether the short-term outcomes of robotic gastrectomy are superior to those of laparoscopic gastrectomy (LG) for elderly patients with advanced gastric cancer has not been reported. METHODS: The study enrolled of 594 elderly patients with advanced gastric cancer who underwent robotic or laparoscopic radical gastrectomy. The RG cohort was matched 1:3 with the LG cohort using propensity score-matching (PSM). RESULTS: After PSM, 121 patients were included in the robot group and 363 patients in the laparoscopic group. Excluding the docking and undocking times, the operation time of the two groups was similar (P = 0.617). The RG group had less intraoperative blood loss than the LG group (P < 0.001). The time to ambulation and first liquid food intake was significantly shorter in the RG group than in the LG group (P < 0.05). The incidence of postoperative complications did not differ significantly between the two groups (P = 0.14). Significantly more lymph nodes were dissected in the RG group than in the LG group (P = 0.001). Postoperative adjuvant chemotherapy was started earlier in the RG group than in the LG group (P = 0.02). CONCLUSIONS: For elderly patients with advanced gastric cancer, RG is safe and feasible. Compared with LG, RG is associated with less intraoperative blood loss; a faster postoperative recovery time, allowing a greater number of lymph nodes to be dissected; and earlier adjuvant chemotherapy.


Subject(s)
Laparoscopy , Robotic Surgical Procedures , Robotics , Stomach Neoplasms , Humans , Aged , Stomach Neoplasms/surgery , Stomach Neoplasms/pathology , Propensity Score , Blood Loss, Surgical , Treatment Outcome , Gastrectomy , Postoperative Complications/surgery , Retrospective Studies
4.
Radiol Med ; 128(6): 644-654, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37148481

ABSTRACT

OBJECTIVE: The objective is to develop a mitotic prediction model and preoperative risk stratification nomogram for gastrointestinal stromal tumor (GIST) based on computed tomography (CT) radiomic features. METHODS: A total of 267 GIST patients from 2009.07 to 2015.09 were retrospectively collected and randomly divided into (6:4) training cohort and validation cohort. The 2D-tumor region of interest was delineated from the portal-phase images on contrast-enhanced (CE)-CT, and radiomic features were extracted. Lasso regression method was used to select valuable features to establish a radiomic model for predicting mitotic index in GIST. Finally, the nomogram of preoperative risk stratification was constructed by combining the radiomic features and clinical risk factors. RESULTS: Four radiomic features closely related to the level of mitosis were obtained, and a mitotic radiomic model was constructed. The area under the curve (AUC) of the radiomics signature model used to predict mitotic levels in training and validation cohorts (training cohort AUC = 0.752; 95% confidence interval [95%CI] 0.674-0.829; validation cohort AUC = 0.764; 95% CI 0.667-0.862). Finally, the preoperative risk stratification nomogram combining radiomic features was equivalent to the clinically recognized gold standard AUC (0.965 vs. 0.983) (p = 0.117). The Cox regression analysis found that the nomogram score was one of the independent risk factors for the long-term prognosis of the patients. CONCLUSION: Preoperative CT radiomic features can effectively predict the level of mitosis in GIST, and combined with preoperative tumor size, accurate preoperative risk stratification can be performed to guide clinical decision-making and individualized treatment.


Subject(s)
Gastrointestinal Stromal Tumors , Humans , Gastrointestinal Stromal Tumors/diagnostic imaging , Gastrointestinal Stromal Tumors/surgery , Mitotic Index , Retrospective Studies , Risk Assessment , Tomography, X-Ray Computed
5.
Radiol Med ; 127(10): 1085-1097, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36057930

ABSTRACT

OBJECTIVE: Development and validation of a radiomics nomogram for predicting recurrence and adjuvant therapy benefit populations in high/intermediate-risk gastrointestinal stromal tumors (GISTs) based on computed tomography (CT) radiomic features. METHODS: Retrospectively collected from 2009.07 to 2015.09, 220 patients with pathological diagnosis of intermediate- and high-risk stratified gastrointestinal stromal tumors and received imatinib treatment were randomly divided into (6:4) training cohort and validation cohort. The 2D-tumor region of interest (ROI) was delineated from the portal-phase images on contrast-enhanced (CE) CT, and radiological features were extracted. The most valuable radiological features were obtained using a Lasso-Cox regression model. Integrated construction was conducted of nomograms of radiomics characteristics to predict recurrence-free survival (RFS) in patients receiving adjuvant therapy. RESULTS: Eight radiomic signatures were finally selected. The area under the curve (AUC) of the radiomics signature model for predicting 3-, 5-, and 7-year RFS in the training and validation cohorts (training cohort AUC = 0.80, 0.84, 0.76; validation cohort AUC = 0.78, 0.80, 0.76). The constructed radiomics nomogram was more accurate than the clinicopathological nomogram for predicting RFS in GIST (C-index: 0.864 95%CI, 0.817-0.911 vs. 0.733 95%CI, 0.675-0.791). Kaplan-Meier survival curve analysis showed a greater benefit from adjuvant therapy in patients with high radiomics scores (training cohort: p < 0.0001; validation cohort: p = 0.017), while there was no significant difference in the low-score group (p > 0.05). CONCLUSION: In this study, a nomogram constructed based on preoperative CT radiomics features could be used for RFS prediction in high/intermediate-risk GISTs and assist the clinical decision-making for GIST patients.


Subject(s)
Gastrointestinal Stromal Tumors , Gastrointestinal Stromal Tumors/diagnostic imaging , Gastrointestinal Stromal Tumors/drug therapy , Humans , Imatinib Mesylate/therapeutic use , Nomograms , Retrospective Studies , Tomography, X-Ray Computed/methods
6.
Surg Endosc ; 36(11): 8639-8650, 2022 11.
Article in English | MEDLINE | ID: mdl-35697854

ABSTRACT

BACKGROUND: Robotic surgery may be advantageous for complex surgery. We aimed to compare the intraoperative and postoperative short-term outcomes of spleen-preserving splenic hilar lymphadenectomy (SPSHL) during robotic and laparoscopic total gastrectomy. METHODS: From July 2016 to December 2020, the clinicopathological data of 115 patients who underwent robotic total gastrectomy combined with robotic SPSHL (RSPSHL) and 697 patients who underwent laparoscopic total gastrectomy combined with laparoscopic SPSHL (LSPSHL) were retrospectively analyzed. A 1:2 ratio propensity score matching (PSM) was used to balance the differences between the two groups to compare their outcomes. The Generic Error Rating Tool was used to evaluate the technical performance. RESULTS: After PSM, the baseline preoperative characteristics of the 115 patients in the RSPSHL and 230 patients in the LSPSHL groups were balanced. The dissection time of the region of the splenic artery trunk (5.4 ± 1.9 min vs. 7.8 ± 3.6 min, P < 0.001), the estimated blood loss during SPSHL (9.6 ± 4.8 ml vs. 14.9 ± 7.8 ml, P < 0.001), and the average number of intraoperative technical errors during SPSHL (15.1 ± 3.4 times/case vs. 20.7 ± 4.3 times/case, P < 0.001) were significantly lower in the RSPSHL group than in the LSPSHL group. The RSPSHL group showed higher dissection rates of No. 10 (78.3% vs. 70.0%, P = 0.104) and No. 11d (54.8% vs. 40.4%, P = 0.012) lymph nodes and significantly improved postoperative recovery results in terms of times to ambulation, first flatus, and first intake (P < 0.05). The splenectomy rates of the two groups were similar (1.7% vs. 0.4%, P = 0.539), and there was no significant difference in morbidity and mortality within postoperative 30 days (13.0% vs. 15.2%, P = 0.589). CONCLUSION: Compared to LSPSHL, RSPSHL has more advantages in terms of surgical qualities and postoperative recovery process with similar morbidity and mortality. For complex SPSHL, robotic surgery may be a better choice.


Subject(s)
Laparoscopy , Robotic Surgical Procedures , Stomach Neoplasms , Humans , Robotic Surgical Procedures/methods , Spleen/surgery , Retrospective Studies , Stomach Neoplasms/surgery , Stomach Neoplasms/pathology , Organ Sparing Treatments/methods , Gastrectomy/methods , Laparoscopy/methods , Lymph Node Excision/methods , Treatment Outcome
7.
Surg Endosc ; 36(11): 8047-8059, 2022 11.
Article in English | MEDLINE | ID: mdl-35764839

ABSTRACT

OBJECTIVE: There is insufficient evidence to evaluate the long-term outcomes of robotic radical gastrectomy. The aim of this study was to compare the radical results and long-term outcomes of robotic and laparoscopic radical gastrectomy. METHODS: We prospectively collected and retrospectively analyzed the general clinicopathological data of gastric cancer patients treated with robotic radical gastrectomy (RG) and laparoscopic radical gastrectomy (LG) from July 2016 to July 2018 at Fujian Medical University Union Hospital. The RG cohort was matched 1:3 with the LG cohort by using propensity score matching (PSM). The primary endpoints of the study were 3-year overall survival (OS) and 3-year relapse-free survival (RFS). RESULTS: The study included 221 patients treated with RG and 1106 patients treated with LG for gastric cancer. After PSM, 211 patients were included in the RG cohort, and 663 patients were included in the LG cohort. The 3-year OS rate was 81.0% in the robotic cohort and 79.3% in the laparoscopic cohort (log-rank test, P = 0.516). The 3-year RFS rate was 78.7% in the robotic cohort and 75.6% in the laparoscopic cohort (log-rank test, P = 0.600). In the subgroup analyses, no significant differences were noted between the RG and LG cohorts in terms of 3-year OS and 3-year RFS (all P > 0.05). The therapeutic value index of each lymph node station dissection in the robotic cohort was comparable to that in the laparoscopic cohort. CONCLUSION: Robotic radical gastrectomy can achieve radical results and long-term outcomes comparable to laparoscopic surgery, and further multicenter prospective studies can be conducted to assess the clinical efficacy of robotic radical gastrectomy.


Subject(s)
Laparoscopy , Robotic Surgical Procedures , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Propensity Score , Retrospective Studies , Prospective Studies , Neoplasm Recurrence, Local/surgery , Gastrectomy/methods , Laparoscopy/adverse effects , Treatment Outcome , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Postoperative Complications/surgery
8.
Food Chem ; 372: 131243, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-34655831

ABSTRACT

High pressure processing (HPP) exhibited different effect on polyphenol oxidase (PPO), but the conformational changes was not clear yet. In this study, molecular dynamics simulation combined with spectroscopic experiments were used to explore PPO conformational changes under high pressure at the molecular level. The simulation results showed that high pressure decreased volume and hydrogen bonds, induced changes in active center and movement of loop. Especially, the conformational changes under 200 and above 400 MPa were different. Under 200 MPa, the distance between His 61 and Cu decreased by 0.4 Å, active pocket was exposed, substrate channel became larger. However, the distance increased by 6.1 Å under 600 MPa, active pocket moved inward, substrate channel became narrower. Docking results of 200 and 600 MPa had the highest and lowest binding affinity, whose T-score was 4.657 and 4.130, respectively. These results were consistent with spectroscopic experiments of PPO after HHP.


Subject(s)
Catechol Oxidase , Molecular Dynamics Simulation , Catechol Oxidase/metabolism , Food Handling , Hot Temperature , Pressure
9.
Drug Deliv ; 28(1): 844-855, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33928829

ABSTRACT

T7 peptide is considered as an antiangiogenic polypeptide. The presents study aimed to further detect the antiangiogenic mechanisms of T7 peptide and determine whether combining T7 peptide and meloxicam (COX-2/PGE2 specific inhibitor) could offer a better therapy to combat hepatocellular carcinoma (HCC). T7 peptide suppressed the proliferation, migration, tube formation, and promoted the apoptosis of endothelial cells under both normoxic and hypoxic conditions via integrin α3ß1 and αvß3 pathways. Cell proliferation, migration, apoptosis, or tube formation ability were detected, and the expression of integrin-associated regulatory proteins was detected. The anti-tumor activity of T7 peptide, meloxicam, and their combination were evaluated in HCC tumor models established in mice. T7 peptide suppressed the proliferation, migration, tube formation, and promoted the apoptosis of endothelial cells under both normoxic and hypoxic conditions via integrin α3ß1 and αvß3 pathways. Meloxicam enhanced the activity of T7 peptide under hypoxic condition. T7 peptide partly inhibited COX-2 expression via integrin α3ß1 not αvß3-dependent pathways under hypoxic condition. T7 peptide regulated apoptosis associated protein through MAPK-dependent and -independent pathways under hypoxic condition. The MAPK pathway was activated by the COX-2/PGE2 axis under hypoxic condition. The combination of T7 and meloxicam showed a stronger anti-tumor effect against HCC tumors in mice. The data highlight that meloxicam enhanced the antiangiogenic activity of T7 peptide in vitro and in vivo.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Collagen Type IV/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/antagonists & inhibitors , Liver Neoplasms/drug therapy , Meloxicam/pharmacology , Peptide Fragments/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation , Cyclooxygenase 2/metabolism , Drug Combinations , Endothelial Cells/drug effects , Humans , Hypoxia/pathology , Integrins/drug effects , Mice , Neovascularization, Pathologic/drug therapy , RNA, Small Interfering/metabolism , Random Allocation , Xenograft Model Antitumor Assays
10.
World J Clin Cases ; 8(10): 2016-2022, 2020 May 26.
Article in English | MEDLINE | ID: mdl-32518795

ABSTRACT

BACKGROUND: Liver infarction is a rare necrotic lesion due to the dual blood supply consisting of the hepatic artery and portal vein. The absence of specific clinical manifestations and imaging appearances usually leads to misdiagnosis and poor prognosis. Thus, the precise diagnosis of liver infarction always requires imaging studies, serum studies, and possible liver biopsy. CASE SUMMARY: We report a case of 31-year-old man who developed a huge liver infarction. Persistent right upper abdominal pain and intermittent fever were the main symptoms in this patient. Computed tomography revealed a huge irregular lesion with a maximum diameter of 12.7 cm in the right lobe of the liver. Three-dimensional reconstruction was performed and no significant interruption of the main hepatic vessels was observed. The lesion was initially considered to be a malignant tumor with internal bleeding. Laparoscopic right hepatectomy was performed, and pathology indicated a rare liver infarction. The patient recovered well and was discharged on postoperative day 21. No fever or abnormal liver function were reported in the subsequent 6 mo. CONCLUSION: In patients with a huge liver infarction, early surgical intervention may be beneficial.

11.
Int J Mol Med ; 44(2): 523-534, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31173192

ABSTRACT

The T7 peptide, an active fragment of full­length tumstatin [the non­collagenous 1 domain of the type IV collagen α3 chain, α3 (IV) NC1], has exhibited potential antitumor effects in several types of cancer cells. However, the mechanism underlying its action against human hepatocellular carcinoma (HCC) remains unclear. The present study aimed to investigate the role of autophagy in T7 peptide­induced cytotoxicity in HCC cells in vitro and in vivo. The results revealed that the T7 peptide significantly reduced cell viability and induced cell cycle arrest in HCC cells. The T7 peptide induced apoptosis in HCC cells through upregulation of Bax, Fas, and Fas ligand, and through upregulation of the anti­apoptotic protein Bcl­2. In addition, treatment with the T7 peptide induced protective autophagy in HCC cells. Blocking autophagy by 3­methyladenineor bafilomycin A1 enhanced T7 peptide­induced apoptosis. Furthermore, co­treatment with MK­2206 (an Akt specific inhibitor) or rapamycin (an inhibitor of mTOR) enhanced T7 peptide­induced autophagy, whereas co­treatment with insulin (an activator of the Akt/mTOR signaling pathway) alleviated T7 peptide­induced autophagy, which suggested that the T7 peptide may induce autophagy activation via inhibition of the Akt/mTOR signaling pathway. Taken together, the present results demonstrated that suppression of autophagy potentiated the cytotoxic effects of the T7 peptide, and suggested that the T7 peptide may serve as a potential alternative compound for HCC therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carcinoma, Hepatocellular/drug therapy , Collagen Type IV/pharmacology , Liver Neoplasms/drug therapy , Peptide Fragments/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Collagen Type IV/therapeutic use , Humans , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Peptide Fragments/therapeutic use
12.
Exp Ther Med ; 17(6): 4628-4634, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31086593

ABSTRACT

The timely and effective treatment for severe acute pancreatitis (SAP) is favorable to prognosis. Decompression of the bile duct might be a feasible way to decrease the progression of SAP. The present study investigated the effects of sustained bile external drainage on organs injury caused by SAP in Sprague-Dawley (SD) rats and the mechanisms involved. A total of 72 female SD rats weighting 190-230 g were randomly divided into four groups (n=18): Sham operation group (SOG), SOG + bile drainage group (BDG), SAP group, and SAP + BDG. Sodium taurocholate solution (4%; 1 mg/kg body weight) was used to set up SAP model via injection of retrograde puncture of biliopancreatic duct through the duodenum. A cannula was inserted into the bile duct and fixed externally to establish BDG model. At each time points (t=3, 6, 12; n=6), tissues from the liver, lung, and pancreas, and blood samples were collected. Serum amylase (AMY) was analyzed in all the samples. The levels of tumor necrosis factor-α (TNF-α), heme oxygenase-1 (HO-1), interleukin-10 (IL-10) and high mobility group box 1 (HMGB1) were detected by ELISA. Hematoxylin-eosin staining was performed to observe the histopathological changes, and nuclear transcription factor (NF)-κB-p65 levels in the pancreas were analyzed by western blotting. The data indicated that BDG alleviated the SAP progression and multiple organs injuries. Meanwhile, the histopathological changes of the pancreas, liver, and lungs were improved by BDG. BDG decreased the pathological scores of pancreas significantly (P<0.05). The levels of AMY, TNF-α, HMGB1, and NF-κB-p65 were significantly downregulated by BDG (P<0.05), while the level of HO-1 was upregulated and IL-10 was unchanged. In summary, BDG may attenuate the multiple organs injuries caused by SAP via downregulation of TNF-α, HMGB1, NF-κB-p65 and upregulation of HO-1.

13.
J Cancer ; 9(8): 1403-1413, 2018.
Article in English | MEDLINE | ID: mdl-29721050

ABSTRACT

Purpose: Systemic therapy has often been used for patients with advanced hepatocellular carcinoma (HCC). However, due to drug resistance, the use of cytotoxic chemotherapy in the treatment of patients with advanced HCC has typically demonstrated low response rates. Secretory clusterin (sCLU) is expressed in aggressive late-stage tumors and associated with resistance to chemotherapy, including that in HCC cases. The present research aimed to investigate the biological role of sCLU in HCC. Methods: sCLU expression in HCC and normal tissues was examined using immunohistochemical staining, followed by analysis of the correlation between sCLU expression and clinical indicators. In addition, the role and internal mechanism of sCLU in cell proliferation and apoptosis were investigated in HCC cells. Results: sCLU expression was significantly upregulated in HCC tissues; and was associated with histological grade and poor overall survival. The levels of sCLU were significantly increased in Bel7402, SMMC7721 and resistant HCC cells (Bel7404-OR). Inhibiting the activity of sCLU enhanced the chemosensitivity of Bel7402 and SMMC7721 cells. Downregulation of sCLU could increase the expression of Gadd45a in HCC cells. Overexpression of sCLU contributed to drug resistance in Bel7402, SMMC7721 and Bel7404-OR cells; whereas, overexpression of Gadd45a alone overcame drug resistance in the cells above. No significant expression changes of sCLU and Gadd45a were observed in HCC cells after the interference of a selective inhibitor of the PI3K/Akt signaling pathway. However, regulation of the expression of Gadd45a could influence the phosphorylation level of Akt; and further regulate the expression of Bcl-2 and Bax proteins involved in the mitochondrial apoptosis pathways. Conclusions: The results demonstrate that sCLU/Gadd45a/PI3K/Akt signaling represents a novel pathway that could regulate drug resistance in a one-way manner in HCC cells.

14.
Oncol Rep ; 38(2): 755-766, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28627623

ABSTRACT

The P18 peptide is a functional fragment of pigment epithelial-derived factor (PEDF), which is an endogenic angiogenesis inhibitor. This study sought to determine the anti-angiogenic bioactivity of the P18 peptide in hepato-cellular carcinoma (HCC) and to elucidate the underlying mechanism. Xenograft tumour growth assays demonstrated the P18 peptide suppressed angiogenesis of HCC in vivo. Wound healing, Transwell and Matrigel-culture assays indicated that the P18 peptide inhibited the cell migration and tube formation of endothelial cells (ECs) in vitro. Cell viability and apoptosis assessed by Cell Counting Kit-8 (CCK-8) and flow cytometry assays suggested that the P18 peptide inhibited angiogenesis by inducing apoptosis of ECs. Angiogenesis- and signal transduction-associated molecules analysed by western blot demonstrated that the P18 peptide targets vascular endothelial cell growth factor receptor 2 (VEGFR2) on ECs. In conclusion, by inhibiting the phosphorylation of VEGFR2, the P18 peptide modulates signalling transduction between VEGF/VEGFR2 and suppresses activation of the PI3K/Akt cascades, leading to an increase in mitochondrial-mediated apoptosis and anti-angiogenic activity. This bioactivity of the P18 peptide may represent a novel therapeutic strategy for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Eye Proteins/genetics , Liver Neoplasms/genetics , Neovascularization, Pathologic/genetics , Nerve Growth Factors/genetics , Serpins/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Apoptosis/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Eye Proteins/therapeutic use , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Mice , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy , Nerve Growth Factors/therapeutic use , Peptides/genetics , Peptides/therapeutic use , Serpins/therapeutic use , Signal Transduction/genetics , Wound Healing/genetics , Xenograft Model Antitumor Assays
15.
Tumour Biol ; 39(5): 1010428317691684, 2017 May.
Article in English | MEDLINE | ID: mdl-28475017

ABSTRACT

Pancreatic ductal adenocarcinoma is one of the most lethal cancers. The Hippo pathway is involved in tumorigenesis and remodeling of tumor microenvironments. Hypoxia exists in the microenvironment of solid tumors, including pancreatic ductal adenocarcinoma and plays a vital role in tumor progression and metastasis. However, it remains unclear how hypoxia interacts with the Hippo pathway to regulate these events. In this study, expressions of yes-associated protein 1 and hypoxia-inducible factor-1α were found to be elevated in pancreatic ductal adenocarcinoma samples compared with those in matched adjacent non-tumor samples. Moreover, hypoxia-inducible factor-1α expression was positively correlated with yes-associated protein 1 level in pancreatic ductal adenocarcinoma tissues. The higher expression of nuclear yes-associated protein 1 was associated with poor histological grade and prognosis for pancreatic ductal adenocarcinoma patients. In vitro, yes-associated protein 1 was highly expressed in pancreatic ductal adenocarcinoma cells. Depletion of yes-associated protein 1 inhibited the invasion of pancreatic ductal adenocarcinoma cells via downregulation of Vimentin, matrix metalloproteinase-2, and matrix metalloproteinase-13, and upregulation of E-cadherin. In addition, hypoxia promoted the invasion of pancreatic ductal adenocarcinoma cells via regulating the targeted genes. Hypoxia also deactivated the Hippo pathway and induced yes-associated protein 1 nuclear translocation. Furthermore, depletion of yes-associated protein 1 or hypoxia-inducible factor-1α suppressed the invasion of pancreatic ductal adenocarcinoma cells under hypoxia. Mechanism studies showed that nuclear yes-associated protein 1 interacted with hypoxia-inducible factor-1α and activated Snail transcription to participate in epithelial-mesenchymal transition-mediated and matrix metalloproteinase-mediated remodeling of tumor microenvironments. Collectively, yes-associated protein 1 is an independent prognostic predictor that interacts with hypoxia-inducible factor-1α to enhance the invasion of pancreatic cancer cells and remodeling of tumor microenvironments. Therefore, yes-associated protein 1 may serve as a novel promising target to enhance therapeutic effects for treating pancreatic cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Phosphoproteins/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Adenocarcinoma/pathology , Adult , Aged , Antigens, CD , Cadherins/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Hypoxia/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 2/genetics , Middle Aged , Phosphoproteins/genetics , Prognosis , Transcription Factors , Tumor Microenvironment , YAP-Signaling Proteins
16.
Cancer Sci ; 108(3): 478-487, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28002618

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive human malignancies. The Yes-associated protein-1 (YAP) plays a critical role in cell proliferation, apoptosis and angiogenesis. Verteporfin is a photosensitizer used in photodynamic therapy and also a small molecular inhibitor of the Hippo-YAP pathway. However, little is known about whether verteporfin could inhibit YAP activity in PDAC cells. Our present results showed that verteporfin suppressed the proliferation of human PDAC PANC-1 and SW1990 cells by arresting cells at the G1 phase, and inducing apoptosis in dose- and time-dependent manners. Verteporfin also inhibited the tumor growth on the PDAC xenograft model. Treatment with verteporfin led to downregulation of cyclinD1 and cyclinE1, modulation of Bcl-2 family proteins and activation of PARP. In addition, verteporfin exhibited an inhibitory effect on angiogenesis and vasculogenic mimicry via suppressing Ang2, MMP2, VE-cadherin, and α-SMA expression in vitro and in vivo. Mechanism studies demonstrated that verteporfin impaired YAP and TEAD interaction to suppress the expression of targeted genes. Our results provide a foundation for repurposing verteporfin as a promising anti-tumor drug in the treatment of pancreatic cancer by targeting the Hippo pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/pathology , DNA-Binding Proteins/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Nuclear Proteins/antagonists & inhibitors , Pancreatic Neoplasms/pathology , Phosphoproteins/antagonists & inhibitors , Porphyrins/pharmacology , Transcription Factors/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, CD , Cadherins/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D1/metabolism , Cyclin E/metabolism , DNA-Binding Proteins/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Phosphoproteins/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , TEA Domain Transcription Factors , Transcription Factors/metabolism , Verteporfin , Vesicular Transport Proteins/antagonists & inhibitors , Xenograft Model Antitumor Assays , YAP-Signaling Proteins , Pancreatic Neoplasms
17.
Oncol Rep ; 35(6): 3614-22, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27109804

ABSTRACT

Recurrence and metastasis are the two leading causes of poor prognosis of hepatocellular carcinoma (HCC) patients. Cyclooxygenase (COX)-2 is overexpressed in many types of cancers including HCC and promotes its metastasis. Meloxicam is a selective COX-2 inhibitor that has been reported to exert an anti-proliferation and invasion/migration response in various tumors. In this study, we examined the role of meloxicam on HCC cell proliferation and migration and explored the molecular mechanisms underlying this effect. We found that meloxicam inhibited HCC cell proliferation and had a cell cycle arrest effect in human HCC cells. Furthermore, meloxicam suppressed the ability of HCC cells expressing higher levels of COX-2 and prostaglandin E2 (PGE2) to migration via potentiating expression of E-cadherin and alleviating expression of matrix metalloproteinase (MMP)-2 and -9. COX-2/PGE2 has been considered to activate the ß-catenin signaling pathway which promotes cancer cell migration. We found that treatment with PGE2 significantly enhanced nuclear accumulation of ß-catenin and the activation of GSK3ß which could be reversed by meloxicam in HCC cells. We also observed that HCC cell migration and upregulation of the level of MMP-2/9 and downregulation of E-cadherin induced by PGE2 were suppressed by FH535, an inhibitor of ß-catenin. Taken together, these findings provide a new treatment strategy against HCC proliferation and migration.


Subject(s)
Cadherins/metabolism , Carcinoma, Hepatocellular/drug therapy , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Liver Neoplasms/drug therapy , Thiazines/pharmacology , Thiazoles/pharmacology , beta Catenin/metabolism , Antigens, CD , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Meloxicam , Neoplasm Invasiveness/pathology , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Sulfonamides/pharmacology , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
18.
Oncol Lett ; 10(2): 778-784, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26622569

ABSTRACT

Sorafenib is a type of multikinase inhibitor that exhibits antiangiogenic and antiproliferative effects; in addition, sorafenib is a unique first-line drug recommended for the treatment of advanced hepatocellular carcinoma (HCC). However, the effectiveness of HCC treatment remains poor due to acquired drug resistance. It has been suggested that hypoxia, induced as a results of the antiangiogenic effects of sustained sorafenib treatment, may be an important factor in sorafenib resistance. The transcription factor hypoxia-inducible factor (HIF)-2α has been reported to be associated with cell proliferation under hypoxic conditions; therefore, it was hypothesized that hypoxia may enhance tumor cell proliferation via this mechanism. The present study aimed to evaluate whether the knock-down of HIF-2α was able to enhance the therapeutic efficacy of sorafenib in order to effectively treat HCC. The results demonstrated that hypoxia protected HCC cells against sorafenib; however, short hairpin RNA-HIF-2α transfection in combination with sorafenib treatment exhibited a significantly synergistic effect against HCC cell proliferation. In addition, HCC cells acquired increased ß-catenin/C-Myc expression, which enhanced proliferation under hypoxic conditions; however, targeted knock-down of HIF-2α or C-Myc markedly decreased cell proliferation in HCC cells. In conclusion, the results of the present study indicated that the targeted knock-down of HIF-2α in combination with sorafenib may be a promising strategy for the treatment of HCC.

19.
Cell Prolif ; 48(6): 691-704, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26481188

ABSTRACT

OBJECTIVES: Meloxicam, a selective cyclooxygenase-2 (COX-2) inhibitor, has been demonstrated to exert anti-tumour effects against various malignancies. However, up to now, mechanisms involved in meloxicam anti-hepatocellular carcinoma effects have remained unclear. MATERIALS AND METHODS: Cell viability and apoptosis were assessed by CCK-8 and flow cytometry. Endoplasmic reticulum (ER) stress and autophagy-associated molecules were analysed by western blotting and immunofluorescence assay. GRP78 and Atg5 knock-down by siRNA or chemical inhibition was used to investigate cytotoxic effects of meloxicam treatment on HCC cells. RESULTS: We found that meloxicam led to apoptosis and autophagy in HepG2 and Bel-7402 cells via a mechanism that involved ER stress. Up-regulation of GRP78 signalling pathway from meloxicam-induced ER stress was critical for activation of autophagy. Furthermore, autophagy activation attenuated ER stress-related cell death. Blocking autophagy by 3-methyladenine (3-MA) or Atg5 siRNA knock-down enhanced meloxicam lethality for HCC by activation of ER stress-related apoptosis. In addition, GRP78 seemed to lead to autophagic activation via the AMPK-mTOR signalling pathway. Blocking AMPK with a chemical inhibitor inhibited autophagy suggesting that meloxicam-regulated autophagy requires activation of AMPK. CONCLUSIONS: Our results revealed that both ER stress and autophagy were involved in cell death evoked by meloxicam in HCC cells. This inhibition of autophagy to enhance meloxicam lethality, suggests a novel therapeutic strategy against HCC.


Subject(s)
Autophagy/drug effects , Carcinoma, Hepatocellular/drug therapy , Endoplasmic Reticulum Stress/drug effects , Liver Neoplasms/drug therapy , Thiazines/pharmacology , Thiazoles/pharmacology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy-Related Protein 5 , Cell Line, Tumor , Cell Survival/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Hep G2 Cells , Humans , Meloxicam , Microtubule-Associated Proteins/genetics , RNA Interference , RNA, Small Interfering
20.
Oncol Rep ; 34(4): 2142-50, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26252057

ABSTRACT

Sorafenib (SOR) is a promising treatment for advanced hepatocellular carcinoma (HCC). However, the precise mechanisms of toxicity and drug resistance have not been fully explored and new strategies are urgently needed for HCC therapy. Meloxicam (MEL) is a selective cyclooxygenase-2 (COX-2) inhibitor which elicits antitumor effects in human HCC cells. In the present study, we investigated the interaction between MEL and SOR in human SMMC­7721 cells and the role endoplasmic reticulum (ER) stress exerts in the combination of SOR with MEL treatment-induced cytotoxicity. Our results revealed that the combination treatment synergistically inhibited cell proliferation and enhanced apoptosis. Furthermore, the combination treatment enhanced ER stress-related molecules which involved in SMMC-7721 cell apoptosis. GRP78 knockdown by siRNA or co-treatment with MG132 significantly increased this combination treatment-induced apoptosis. In addition, we found that the combination treatment suppressed tumor growth by way of activation of ER stress in in vivo models. We concluded that the combination of SOR with MEL treatment-induced ER stress, and eventually apoptosis in human SMMC-7721 cells. Knockdown of GRP78 using siRNA or proteosome inhibitor enhanced the cytotoxicity of the combination of SOR with MEL-treatment in SMMC-7721 cells. These findings provided a new potential treatment strategy against HCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Endoplasmic Reticulum Stress/drug effects , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Thiazines/administration & dosage , Thiazoles/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/metabolism , Meloxicam , Mice , Niacinamide/administration & dosage , Niacinamide/metabolism , Phenylurea Compounds/metabolism , Sorafenib , Thiazines/pharmacology , Thiazoles/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...