Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Lett ; 558: 216059, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36813650

ABSTRACT

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief and Authors. Following concerns raised in the public domain, the authors contacted the journal to request the retraction of the article. Sections of panels from various figures appear similar to each other, particularly panels from Figs. 3G, 5B and 3G and 5F, 3F, S4D, S5D, S5C and S10C, as well as S10E.

2.
Gut ; 72(9): 1722-1737, 2023 09.
Article in English | MEDLINE | ID: mdl-36828627

ABSTRACT

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal tumour with limited treatment options. Here, we identified syndecan binding protein (SDCBP), also known as syntenin1, as a novel targetable factor in promoting PDAC tumour progression. We also explored a therapeutic strategy for suppressing SDCBP expression. DESIGN: We used samples from patients with PDAC, human organoid models, LSL-KrasG12D/+mice, LSL-Trp53R172H/+ and Pdx1-Cre (KPC) mouse models, and PDX mouse models. Immunostaining, colony formation assay, ethynyl-2-deoxyuridine incorporation assay, real-time cell analysis, cell apoptosis assay, automated cell tracking, invadopodia detection and gelatin degradation assays, coimmunoprecipitation, and pull-down assays were performed in this study. RESULTS: The median overall survival and recurrence-free survival rates in the high-SDCBP group were significantly shorter than those in the low-SDCBP group. In vitro and in vivo studies have demonstrated that SDCBP promotes PDAC proliferation and metastasis. Mechanically, SDCBP inhibits CK1δ/ε-mediated YAP-S384/S387 phosphorylation, which further suppresses ß-TrCP-mediated YAP1 ubiquitination and proteasome degradation by directly interacting with YAP1. SDCBP interacts with the TAD domain of YAP1, mainly through its PDZ1 domain. Preclinical KPC mouse cohorts demonstrated that zinc pyrithione (ZnPT) suppresses PDAC tumour progression by suppressing SDCBP. CONCLUSIONS: SDCBP promotes the proliferation and metastasis of PDAC by preventing YAP1 from ß-TrCP-mediated proteasomal degradation. Therefore, ZnPT could be a promising therapeutic strategy to inhibit PDAC progression by suppressing SDCBP.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Mice , Animals , beta-Transducin Repeat-Containing Proteins/metabolism , Pancreatic Neoplasms/pathology , Pancreas/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Syntenins/metabolism , Pancreatic Neoplasms
3.
Int J Biol Sci ; 18(3): 911-922, 2022.
Article in English | MEDLINE | ID: mdl-35173526

ABSTRACT

Pancreatic cancer is a dismal malignancy with poor prognosis. In spite of progress in surgical technology, chemotherapy is still the cornerstone in the multi-disciplinary treatment. Albumin-bound paclitaxel is a first-line treatment for PDAC patients. Yet the response rate of the drug is far from satisfying. SOX8 is a member of the sex determining region Y-boxes family, which is potentially related to the chemoresistance of tumor. Patient with high expression of SOX8 were insensitive to albumin-bound paclitaxel. SOX8 reduced apoptosis and G2/M cell cycle arrest caused by albumin-bound paclitaxel. SOX8 transcriptionally regulated EZH2, which reduced expression of SPARC by promoting the methylation of SPARC, thereby reducing the transport of albumin-bound paclitaxel in pancreatic cancer cells. EZH2 inhibitor, UNC1999, can reverse the effect of SOX8 on chemo-resistance of albumin-bound paclitaxel. Collectively, our data revealed SOX8/EZH2/SPARC signaling induced primary chemo-resistance of albumin-bound paclitaxel in pancreatic ductal adenocarcinoma.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Albumin-Bound Paclitaxel/metabolism , Albumin-Bound Paclitaxel/pharmacology , Albumin-Bound Paclitaxel/therapeutic use , Apoptosis/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Osteonectin/genetics , Osteonectin/pharmacology , Osteonectin/therapeutic use , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Pancreatic Neoplasms/metabolism , SOXE Transcription Factors/metabolism , Pancreatic Neoplasms
4.
Thorac Cancer ; 13(3): 369-379, 2022 02.
Article in English | MEDLINE | ID: mdl-34951127

ABSTRACT

BACKGROUND: The purpose of this study was to explore the reason for guanine nucleotide binding-protein gamma subunit-4 (GNG4) overexpression and the relationship between GNG4 overexpression and the poor prognosis of lung adenocarcinoma (LUAD) patients. METHODS: The genes and phenotypes related to GNG4 expression in patients with lung adenocarcinoma were analyzed by bioinformatics. The phenotype indicated by bioinformatic analysis was confirmed by experiments. RESULTS: GNG4 expression is elevated in lung adenocarcinoma, and overexpressed GNG4 is related to the poor prognosis of patients with lung adenocarcinoma. The hypoxic microenvironment of lung adenocarcinoma can promote GNG4 expression and GNG4 promotes the migration and proliferation of lung adenocarcinoma cells. CONCLUSIONS: GNG4 expression in lung adenocarcinoma was significantly higher than in paired adjacent tissues. GNG4 overexpression is associated with a variety of malignant phenotypes of lung adenocarcinoma. Increased GNG4 expression is related to the hypoxic microenvironment in lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Adenocarcinoma of Lung/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Prognosis , Tumor Microenvironment
5.
Pancreatology ; 21(4): 763-770, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33712388

ABSTRACT

BACKGROUND: FOLFIRINOX and gemcitabine plus albumin-bound paclitaxel (AG) regimens are recommended as first-line therapy for both locally advanced and metastatic pancreatic cancer. However, there were no specific markers to conduct personalized regimen choice. The research is to assess delta Housfield unit (delta HU), which is the difference in CT attenuation value (in HU) between enhanced and nonenhanced phase of region of interest, as a marker for predicting chemotherapy response of unresectable pancreatic cancer. METHODS: A total of 179 unresectable pancreatic cancer patients were enrolled in the study. Kaplan-Meier analysis and COX regression analysis were performed for progression-free survival (PFS) and overall survival. The differences of clinical characteristics were analyzed by χ 2test. Microvessel density (MVD) was calculated by immunochemistry staining of CD34. RESULTS: Delta HU was an independent risk factor for unresectable pancreatic cancer (P = 0.017, HR 0.672, 95%CI 0.485-0.930). Patients with higher delta HU were associated with better PFS (P = 0.004). For modified FOLFIRINOX (mFOLFIRINOX) group, delta HU was an independent risk factor (P = 0.045, HR 0.571), but not for AG group (P = 0.473, HR 0.855). Delta HU was correlated with stroma MVD (P = 0.000, R = 0.483), not with parenchyma MVD (P = 0.074, R = 0.199). CONCLUSIONS: Delta HU was a marker predicting chemotherapy response for unresectable pancreatic cancer. Higher delta HU was associated with better survival for patients receiving mFOLFIRINOX rather than AG. The delta HU was positively correlated with stroma MVD, explaining the relationship between delta HU and prognosis.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy , Prognosis , Pancreatic Neoplasms
6.
Thorac Cancer ; 10(8): 1702-1709, 2019 08.
Article in English | MEDLINE | ID: mdl-31268619

ABSTRACT

BACKGROUND: ANLN has been identified as an actin-binding protein and previous studies have suggested that ANLN is associated with actin cytoskeletal dynamics. Lung adenocarcinoma is a poor prognosis tumor. Metastasis is a very common complication and is regarded as the main cause for an unsatisfactory treatment outcome. Whether ANLN is involved in the metastasis of lung adenocarcinoma is unknown. METHODS: We performed immunohistochemical staining and analyzed the correlation between the expression level and pathological parameters. We tested the migration and invasion of A549 and PC9 cell after interference with ANLN expression by Transwell and scratch wound healing assays. Protein expression of E-cadherin, vimentin and N-cadherin were detected using Western blotting and immunofluorescence staining. The same experiment was also tested after overexpression of ANLN. RESULTS: The metastasis of patients with high expression of ANLN was significantly more than that of patients with low expression of ANLN. In vitro, after interfering with ANLN expression, E-cadherin and vimentin expression were increased and N-cadherin expression was decreased in A549 and PC9 cells. Migration and invasion ability of A549 and PC9 cells were decreased,vice versa. CONCLUSION: Our study suggests that the expression of ANLN in lung adenocarcinoma is associated with metastasis of cancer cells. ANLN may be involved in the metastasis of lung adenocarcinoma by promoting epithelial mesenchymal transformation of tumor cells.


Subject(s)
Adenocarcinoma of Lung/metabolism , Lung Neoplasms/metabolism , Microfilament Proteins/biosynthesis , A549 Cells , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Middle Aged , Neoplasm Metastasis , Transfection
7.
Cancer Lett ; 453: 10-20, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30922919

ABSTRACT

Perineural invasion (PNI) is a typical pathological feature of pancreatic ductal adenocarcinoma (PDAC). PNI is associated with poor prognosis of PDAC patients, however, the underlying molecular mechanisms in the development of PNI have not been fully revealed. In this study, we found that the expression of GM-CSF and HIF-1α were dramatically increased in PDAC cells. The overexpression of HIF-1α and GM-CSF closely correlated with increased occurrence of PNI and cancer-related pain and shortened overall survival in PDAC patients. GM-CSF expression in PDAC cells was mediated by HIF-1α through direct binding to the hypoxia response element in the GM-CSF promoter. The activated HIF-1α can up-regulate GM-CSF expression and secretion, which promoted the migration of Schwann cells in tumor microenvironment. Furthermore, GM-CSF overexpression could rescue the inhibition of Schwann cells migration by HIF-1α knockdown. Moreover, HIF-1α inhibition with PX478 drastically reduced the expression level of GM-CSF and decreased the PNI in a PDAC xenograft mouse model. Overall, our results demonstrated that the HIF-1α/GM-CSF pathway is involved in the tumor-nerve interactions and promotes the occurrence of PNI. The blockade of this signal might help to inhibit PDAC progression.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pancreatic Neoplasms/pathology , Schwann Cells/pathology , Adult , Aged , Animals , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Movement/physiology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Pancreatic Neoplasms/metabolism , Peripheral Nerves/pathology , Schwann Cells/metabolism , Up-Regulation
8.
Clin Sci (Lond) ; 133(5): 645-663, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30782607

ABSTRACT

Forkhead box protein M1 (FOXM1) was identified as an oncogenic transcription factor and master regulator of tumor progression and metastasis. FOXM1 expression often correlates with poor prognosis and chemotherapy resistance. In the present study, we investigated the association of FOXM1 expression and chemoresistance in pancreatic cancer. Elevated FOXM1 protein levels were associated with gemcitabine chemoresistance in patients with pancreatic cancer. In gemcitabine resistance cell line models of pancreatic cancer, FOXM1 expression increased, which induced gemcitabine chemoresistance in vitro In pancreatic cancer cells treated with gemcitabine, FOXM1 affected nuclear factor κB (NF-κB) signaling activity. Immunohistochemical analysis demonstrated a negative association of FOXM1 expression and the level of phosphorylated signal transducer and activator of transcription 1 (pSTAT1) in human pancreatic cancer tissues. Dual-luciferase reporter assays and chromatin-immunoprecipitation assays demonstrated that pSTAT1 directly binds to the FOXM1 promoter to down-regulate its transcription. Interferon γ (IFNγ) promoted gemcitabine-induced cell apoptosis and inhibited cell proliferation in vitro and in vivo by FOXM1 inhibition. These data suggested that FOXM1 enhances chemoresistance to gemcitabine in pancreatic cancer. IFNγ could be used to down-regulate the expression of FOXM1 through STAT1 phosphorylation, thereby increasing the sensitivity of pancreatic cancer cells to gemcitabine. These studies suggested the sensitization by IFNγ in pancreatic ductal adenocarcinoma (PDAC) chemotherapy, which requires further clinical studies.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Forkhead Box Protein M1/metabolism , Interferon-gamma/pharmacology , Pancreatic Neoplasms/drug therapy , STAT1 Transcription Factor/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Forkhead Box Protein M1/genetics , Humans , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
9.
Thorac Cancer ; 9(10): 1235-1240, 2018 10.
Article in English | MEDLINE | ID: mdl-30070037

ABSTRACT

BACKGROUND: The purpose of the study was to investigate the association between radiomic features based on contrast-enhanced multidetector computed tomography (CT) and the Ki-67 proliferation index (PI) in patients with lung cancer. METHODS: One hundred and ten patients with lung cancer confirmed by surgical histology were retrospectively included. Radiomic features were extracted from preoperative contrast-enhanced chest multidetector CT images for each tumor using open-source three-dimensional Slicer software. Statistical analysis was performed to determine significant radiomic features serving as image predictors of Ki-67 status in lung cancer and to investigate the relationship between these features and Ki-67 PI. RESULTS: Higher Ki-67 expression was more common in men (P = 0.02) and patients with a smoking history (P = 0.01). Twelve radiomic features were significantly associated with Ki-67 status. Multivariate logistic regression analysis identified inverse variance, minor axis, and elongation as independent predictors of Ki-67 PI. There was a positive correlation between inverse variance, minor axis, elongation (P = 0.00, P = 0.02, and P = 0.14, respectively) and Ki-67 PI. The area under the curve to identify high Ki-67 status for inverse variance was 0.77 with a cutoff value of 0.47, which was significantly higher than for minor axis and elongation (P = 0.02 and P = 0.03, respectively). CONCLUSION: Radiomic features based on contrast CT images, including inverse variance, minor axis, and elongation, can serve as noninvasive predictors of Ki-67 status in patients with lung cancer. Inverse variance could be superior to the other radiomic features to identify high Ki-67 status.


Subject(s)
Ki-67 Antigen/metabolism , Lung Neoplasms/diagnostic imaging , Tomography, X-Ray Computed/methods , Adult , Aged , Cell Proliferation , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...