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










Publication year range
1.
Breast Cancer Res ; 26(1): 1, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167446

ABSTRACT

BACKGROUND: Despite radiotherapy ability to significantly improve treatment outcomes and survival in triple-negative breast cancer (TNBC) patients, acquired resistance to radiotherapy poses a serious clinical challenge. Protein disulfide isomerase exists in endoplasmic reticulum and plays an important role in promoting protein folding and post-translational modification. However, little is known about the role of protein disulfide isomerase family member 4 (PDIA4) in TNBC, especially in the context of radiotherapy resistance. METHODS: We detected the presence of PDIA4 in TNBC tissues and paracancerous tissues, then examined the proliferation and apoptosis of TNBC cells with/without radiotherapy. As part of the validation process, xenograft tumor mouse model was used. Mass spectrometry and western blot analysis were used to identify PDIA4-mediated molecular signaling pathway. RESULTS: Based on paired clinical specimens of TNBC patients, we found that PDIA4 expression was significantly higher in tumor tissues compared to adjacent normal tissues. In vitro, PDIA4 knockdown not only increased apoptosis of tumor cells with/without radiotherapy, but also decreased the ability of proliferation. In contrast, overexpression of PDIA4 induced the opposite effects on apoptosis and proliferation. According to Co-IP/MS results, PDIA4 prevented Tax1 binding protein 1 (TAX1BP1) degradation by binding to TAX1BP1, which inhibited c-Jun N-terminal kinase (JNK) activation. Moreover, PDIA4 knockdown suppressed tumor growth xenograft model in vivo, which was accompanied by an increase in apoptosis and promoted tumor growth inhibition after radiotherapy. CONCLUSIONS: The results of this study indicate that PDIA4 is an oncoprotein that promotes TNBC progression, and targeted therapy may represent a new and effective anti-tumor strategy, especially for patients with radiotherapy resistance.


Subject(s)
MAP Kinase Signaling System , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Disulfide-Isomerases/pharmacology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/radiotherapy , Triple Negative Breast Neoplasms/drug therapy , Carcinogenesis , Cell Transformation, Neoplastic , Family , Cell Line, Tumor , Cell Proliferation
2.
Int J Med Sci ; 20(12): 1616-1630, 2023.
Article in English | MEDLINE | ID: mdl-37859699

ABSTRACT

Purpose: Acute liver failure (ALF) is a clinically fatal disease that leads to the rapid loss of normal liver function. Acetaminophen (APAP) is a leading cause of drug-induced ALF. Ferroptosis, defined as iron-dependent cell death associated with lipid peroxide accumulation, has been shown to be strongly associated with APAP-induced liver injury. Growth arrest-specific 1 (GAS1) is a growth arrest-specific gene, which is closely related to the inhibition of cell growth and promotion of apoptosis. However, the functional role and underlying mechanism of GAS1 in APAP-induced ferroptosis remain unknown. Methods: We established liver-specific overexpression of GAS1 (GAS1AAV8-OE) mice and the control (GAS1AAV8-vector) mice by tail vein injection of male mice with adeno-associated virus. APAP at 500 mg/kg was intraperitoneally injected into these two groups of mice to induce acute liver failure. The shRNA packaged by the lentivirus inhibits GAS1 gene expression in human hepatoma cell line HepaRG (HepaRG-shNC and HepaRG-shGAS1-2) and primary hepatocytes of mice with liver-specific overexpression of GAS1 were isolated and induced by APAP in vitro to further investigate the regulatory role of GAS1 in APAP-induced acute liver failure. Results: APAP-induced upregulation of ferroptosis, levels of lipid peroxides and reactive oxygen species, and depletion of glutathione were effectively alleviated by the ferroptosis inhibitor, ferrostatin-1, and downregulation of GAS1 expression. GAS1 overexpression promoted ferroptosis-induced lipid peroxide accumulation via p53, inhibiting its downstream target, solute carrier family 7 member 11. Conclusion: Collectively, our findings suggest that GAS1 overexpression plays a key role in aggravating APAP-induced acute liver injury by promoting ferroptosis-induced accumulation of lipid peroxides.


Subject(s)
Ferroptosis , Liver Failure, Acute , Animals , Humans , Male , Mice , Acetaminophen/toxicity , Cell Cycle Proteins/metabolism , Ferroptosis/genetics , GPI-Linked Proteins/metabolism , Hepatocytes/metabolism , Lipid Peroxides/metabolism , Liver , Liver Failure, Acute/chemically induced , Liver Failure, Acute/genetics , Liver Failure, Acute/metabolism , Mice, Inbred C57BL
3.
ACS Appl Mater Interfaces ; 15(37): 44305-44313, 2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37698376

ABSTRACT

The compelling integration of superhydrophobic coatings with light-to-heat conversion capabilities has garnered substantial interest due to their dual functionality encompassing passive anti-icing and deicing attributes. However, the insufficient mechanical stability and the environmental and human health concerns stemming from the extensive use of organic solvents limit their practical application. In this study, an all-waterborne superhydrophobic photothermal coating (PCPAS) was prepared through the synergy of composite micro-nanoparticles derived from carbon nanotubes (CNT), polydopamine (PDA), and Ag particles with fluorine-containing polyacrylic emulsion (PFA). The PDA provided active sites for Ag+ reduction reaction and enhanced the interfacial interaction between CNT and Ag particles. The interfacial enhancement enabled the coating to maintain stable superhydrophobicity after 260 times sandpaper abrasion and 240 times tape peeling. Simultaneously, the composite micro-nanoparticle's light-to-heat conversion ability gave the coating excellent anti-icing/deicing capabilities. Under the condition of -20 °C, the freezing time of 30 µL of water droplets was extended to 392 s, and 2 × 2 × 2 cm ice cubes placed on the surface of the coating could completely melt after only 1142 s under simulated sunlight irradiation with a 1 kW/m2 intensity. In addition, the coating also had suitable self-cleaning properties and substrate applicability. The comprehensive attributes of this all-waterborne photothermal superhydrophobic coating render it a promising contender for anti-icing and deicing applications in challenging outdoor environments.

4.
Am J Cancer Res ; 13(8): 3500-3516, 2023.
Article in English | MEDLINE | ID: mdl-37693125

ABSTRACT

Insulinoma-associated protein-1 (INSM1), which is highly expressed in various neuroendocrine tumors, functions as a zinc finger transcription factor capable of regulating the biological behavior of tumor cells. However, its specific role in breast cancer remains unclear. This study aims to investigate the role and mechanism of INSM1 in breast cancer. A total of 158 cohorts were recruited to examine the expression of INSM1 in breast cancer tissues and their corresponding adjacent normal tissues using immunohistochemistry. Follow-up data, along with clinical and pathological information, were collected to analyze the correlation between INSM1 expression and survival outcomes in breast cancer patients. Additionally, we investigated the impact of INSM1 on breast cancer cell proliferation, migration, and aggregation. To further explore the regulatory effect of INSM1 knockdown on breast cancer tumor growth, we utilized a xenograft mouse model. The results revealed that INSM1 was significantly overexpressed in breast cancer patients and correlated with prognosis. Knockdown of INSM1 notably impaired the malignant biological effects of breast cancer cells and inhibited the growth of xenograft tumors in nude mice. Importantly, our data also suggests an interaction between INSM1 and S-phase kinase-associated protein 2 (SKP2), which in turn regulates C-MYC, thereby affecting the p-ERK pathway. Our study provides the first evidence demonstrating the contribution of INSM1 to tumor formation and growth in breast cancer. Furthermore, we found that INSM1 positively regulates C-MYC and the p-ERK pathway by interacting with SKP2 during breast cancer development. Collectively, these findings highlight INSM1 as a promising target for breast cancer treatment.

5.
J Cell Physiol ; 235(12): 9388-9398, 2020 12.
Article in English | MEDLINE | ID: mdl-32342525

ABSTRACT

Kinesin family member 15 (KIF15) is a member of the kinesin superfamily of proteins, which promotes cell mitosis, participates in the transport of intracellular materials, and helps structural assembly and cell signaling pathways transduction. However, its biological role and molecular mechanisms of action in the development of gastric cancer (GC) remain unclear. In the present study, an integrated analysis of The Cancer Genome Atlas (TCGA), Gene Expression Omnibus database, and Kaplan-Meier plotter database was performed to predict the expression and prognostic value of KIF15 in GC patients. Detection of KIF15 expression in GC cells and tissues was performed by a quantitative polymerase chain reaction. In vitro cell proliferation, viability, colony formation ability and flow cytometry assays, and in vivo tumorigenicity assay, were performed to evaluate the effects of KIF15 knockdown on GC cell phenotype. It was demonstrated that the expression of KIF15 messenger RNA in GC tissues was significantly higher compared with that in adjacent tissues, and was closely associated with larger tumor size and poor patient prognosis. In addition, functional studies demonstrated that, due to the increase in reactive oxygen species (ROS) generation, the interference with the expression of KIF15 not only decreased cell proliferation but also increased cell apoptosis and induced cell cycle arrest. ROS-mediated activation of c-Jun N-terminal kinase/c-Jun signaling reduced cell proliferation by regulating the GC cell cycle and increasing apoptosis. Taken together, the results of the present study indicate that KIF15 is an oncoprotein contributing to GC progression, and is expected to help identify novel biomarkers and treatment targets in GC.


Subject(s)
Apoptosis/genetics , Kinesins/genetics , Reactive Oxygen Species/metabolism , Stomach Neoplasms/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kinesins/metabolism , Male , Mice , Middle Aged , Stomach Neoplasms/genetics
6.
Medicine (Baltimore) ; 99(13): e19539, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32221073

ABSTRACT

Gallbladder occupying lesions are common diseases of biliary system. Among them, gallbladder cancer is difficult to diagnose due to the indistinguishable early symptoms, thus posing a great risk to the population. This study aims to establish a computed tomography (CT) prediction model for distinguishing benign and malignant lesions of gallbladder occupying lesions.The study included 211 patients with benign or malignant gallbladder occupying lesions who have taken resection in the Nanjing Drum Tower Hospital from January 2009 to December 2017. Clinical data collected includes age and sex; CT data includes tumor location, tumor maximum diameter, tumor form, venous phase portal venous CT value, abdominal aortic CT value, plain phase CT value, arterial phase CT value, venous phase CT value, delayed phase CT value, ΔCT1, ΔCT2, ΔCT3, ΔCT4, ΔCT5, ΔCT6, and ΔCT7. Calculation of odds ratio between benign and malignant gallbladder occupying lesions using single factor screening variables and multivariate logistic regression was done to establish a model and calculate the areas under receiver operating characteristic curves of the model.Multivariate logistic regression analysis showed that age, tumor maximum diameter, tumor form, venous phase portal venous CT value, ΔCT2, ΔCT4, and ΔCT6 are the main characteristic index for differential diagnosis of benign and malignant risk of gallbladder occupying lesions.Patients' age, tumor maximum diameter, tumor form, venous phase portal venous CT value, ΔCT2, ΔCT4, and ΔCT6 are independent risk factors for judging the benign and malignant of gallbladder occupying lesions. The model established exhibited a potential diagnostic value for distinguishing the malignant properties of gallbladder occupying lesions.


Subject(s)
Gallbladder Diseases/diagnostic imaging , Gallbladder Diseases/pathology , Tomography, X-Ray Computed/methods , Adult , Age Factors , Aged , Contrast Media , Diagnosis, Differential , Female , Gallbladder Diseases/diagnosis , Gallbladder Neoplasms/diagnostic imaging , Gallbladder Neoplasms/pathology , Humans , Male , Middle Aged , Regression Analysis , Sex Factors , Tumor Burden
7.
Nanomedicine (Lond) ; 14(19): 2579-2593, 2019 10.
Article in English | MEDLINE | ID: mdl-31609675

ABSTRACT

Aim: To investigate the role of exosomal miRNAs on gastric cancer (GC) metastasis. Materials & methods: miRNA expression profiles of exosomes with distinct invasion potentials were analyzed using miRNA microarray and validated by quantitative real-time PCR. In vitro and in vivo experiments assessed the role of exosomal miR-196a-1 in GC's metastasis. Results: High expression level of exosomal miR-196a-1 expression was significantly associated with poor survival in GC. Exosomes that contained miR-196a-1 were secreted from high-invasive GC cells. Ectopic miR-196a-1 expression promoted invasion of low-invasive GC cells by targeting SFRP1. Conclusion: miR-196a-1 was delivered from high-invasive GC into low-invasive GC cells via exosomes and promoted metastasis to the liver in vitro and in vivo.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Stomach Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Coculture Techniques , Exosomes/genetics , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Mice , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Stomach Neoplasms/pathology
8.
Int J Biol Sci ; 15(11): 2320-2329, 2019.
Article in English | MEDLINE | ID: mdl-31595150

ABSTRACT

Solid tumors consist of various types of stromal cells in addition to cancer cells. Cancer-associated fibroblasts (CAFs) are a major component of the tumor stroma and play an essential role in tumor progression and metastasis in a variety of malignancies, including gastric cancer. However, the effects of CAFs on gastric cancer cells' progression and metastasis are not well studied. Here we show that matrix metalloproteinase 11 (MMP11) in exosomes secreted from CAFs can be delivered into gastric cancer cells. Gastric CAFs promote gastric cancer cell migration partially through exosomal MMP11. Moreover, MMP11 is overexpressed in exosomes purified from plasma of gastric cancer patients and tumor tissues and associated with overall survival of gastric patients. We also find that MMP11 is negatively regulated by exosomal miR-139 in the CAFs of gastric cancer. Exosomal miR-139 inhibits tumor growth and metastasis of gastric cancer cells by decreasing the expression of MMP11 in vitro and in vivo. Thus, we propose that exosomal miR-139 derived from gastric CAFs could inhibit the progression and metastasis of gastric cancer by decreasing MMP11 in tumor microenvironment.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Exosomes/genetics , Matrix Metalloproteinase 11/genetics , MicroRNAs/metabolism , Stomach Neoplasms/genetics , Animals , Cancer-Associated Fibroblasts/enzymology , Cell Movement/genetics , Disease Progression , Exosomes/enzymology , Exosomes/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 11/metabolism , Mice, Nude , Neoplasm Metastasis , Stomach Neoplasms/enzymology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
9.
Cell Death Dis ; 7(12): e2569, 2016 12 29.
Article in English | MEDLINE | ID: mdl-28032859

ABSTRACT

Epigenetic silencing of tumor suppressors contributes to the development and progression of colorectal cancer (CRC). We recently found that speckle-type POZ protein (SPOP) was significantly downregulated and the inactivation of SPOP promoted metastasis in CRC. This study aimed to clarify its epigenetic alteration, molecular mechanisms and clinical significance in CRC. Our results revealed that the core region of SPOP promoter was hypermethylated in CRC tissues and its methylation was correlated with poor survival. Transcription factor RXRA had a vital role in the regulation of SPOP gene. The data indicated that DNA methylation at -167 bp of the SPOP gene altered the binding affinity between transcription factor RXRA and SPOP promoter. Moreover, SPOP was found to associate with Gli2 and promoted its ubiquitination and degradation in CRC. Consequently, the expression level of Hh/Gli2 pathway-related apoptotic protein Bcl-2 was decreased and the function of resisting cell death was inhibited in CRC. It suggests that methylation status of SPOP promoter can be used as a novel epigenetic biomarker and a therapeutic target in CRC.


Subject(s)
Apoptosis/genetics , Colorectal Neoplasms/genetics , DNA Methylation/genetics , Gene Silencing , Hedgehog Proteins/metabolism , Nuclear Proteins/genetics , Promoter Regions, Genetic , Repressor Proteins/genetics , Up-Regulation/genetics , Aged , Animals , Anoikis/genetics , Cell Proliferation/genetics , CpG Islands/genetics , Female , Humans , Kruppel-Like Transcription Factors/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Nuclear Proteins/metabolism , Protein Binding/genetics , Proteolysis , Repressor Proteins/metabolism , Retinoid X Receptor alpha/metabolism , Signal Transduction/genetics , Transcription, Genetic , Ubiquitination , Zinc Finger Protein Gli2
10.
Free Radic Biol Med ; 99: 593-607, 2016 10.
Article in English | MEDLINE | ID: mdl-27634171

ABSTRACT

Natriuretic peptide receptor A (NPRA), the major receptor for atrial natriuretic peptide (ANP), has been implicated in tumorigenesis; however, the role of ANP-NPRA signaling in the development of gastric cancer remains unclear. Immunohistochemical analyses indicated that NPRA expression was positively associated with gastric tumor size and cancer stage. NPRA inhibition by shRNA induced G2/M cell cycle arrest, cell death, and autophagy in gastric cancer cells, due to accumulation of reactive oxygen species (ROS). Either genetic or pharmacologic inhibition of autophagy led to caspase-dependent cell death. Therefore, autophagy induced by NPRA silencing may represent a cytoprotective mechanism. ROS accumulation activated c-Jun N-terminal kinase (JNK) and AMP-activated protein kinase (AMPK). ROS-mediated activation of JNK inhibited cell proliferation by disturbing cell cycle and decreased cell viability. In addition, AMPK activation promoted autophagy in NPRA-downregulated cancer cells. Overall, our results indicate that the inhibition of NPRA suppresses gastric cancer development and targeting NPRA may represent a promising strategy for the treatment of gastric cancer.


Subject(s)
Autophagy/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , Reactive Oxygen Species/agonists , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Acetylcysteine/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Anthracenes/pharmacology , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Autophagy/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , G2 Phase Cell Cycle Checkpoints/genetics , Humans , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Nude , Neoplasm Staging , Pyridines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
J Exp Clin Cancer Res ; 34: 139, 2015 Nov 14.
Article in English | MEDLINE | ID: mdl-26576539

ABSTRACT

BACKGROUND: The receptor tyrosine kinase-like orphan receptors (ROR) family contains the atypical member ROR1, which plays an oncogenic role in several malignant tumors. However, the clinical potentials and underlying mechanisms of ROR1 in gastric cancer progression remain largely unknown. In this study, we validated the microRNA-mediated gene repression mechanism involved in the role of ROR1. METHODS: Bioinformatic prediction, luciferase reporter assay, quantitative real-time PCR (qRT-PCR) and western blotting were used to reveal the regulatory relationship between miR-27b-3p and ROR1. The expression patterns of miR-27b-3p and ROR1 in human gastric cancer (GC) specimens and cell lines were determined by microRNA RT-PCR and western blotting. Cell proliferation, colony formation assay in soft agar in vitro and tumorigenicity in vivo were performed to observe the effects of downregulation and upregulation miR-27b-3p expression on GC cell phenotypes. RESULTS: miR-27b-3p suppressed ROR1 expression by binding to the 3'UTR of ROR1 mRNA in GC cells. miR-27b-3p was significantly downregulated and reversely correlated with ROR1 protein levels in clinical samples. Analysis of the clinicopathological significance showed that miR-27b-3p and ROR1 were closely correlated with GC characteristics. Ectopic miR-27b-3p expression suppressed cell proliferation, colony formation in soft agar, xenograft tumors of GC cells. By contrast, miR-27b-3p knockdown enhanced these malignant behaviors. Our studies further revealed that the c-Src/STAT3 signaling pathway was involved in miR-27b-3p-ROR1-mediated cell proliferation regulation. CONCLUSIONS: These results show that miR-27b-3p suppresses ROR1 expression through the binding site in the 3'UTR inhibiting the cell proliferation. These findings indicate that miR-27b-3p exerts tumor-suppressive effects in GC through the suppression of oncogene ROR1 expression and suggest a therapeutic application of miR-27b-3p in GC.


Subject(s)
MicroRNAs/genetics , RNA Interference , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Stomach Neoplasms/genetics , 3' Untranslated Regions , Adult , Aged , Animals , Base Sequence , Binding Sites , CSK Tyrosine-Protein Kinase , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , MicroRNAs/chemistry , Middle Aged , Neoplasm Grading , Neoplasm Staging , RNA, Messenger/chemistry , RNA, Messenger/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Burden , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
12.
Cell Physiol Biochem ; 35(2): 571-85, 2015.
Article in English | MEDLINE | ID: mdl-25612651

ABSTRACT

BACKGROUND: A proliferation-inducing ligand (APRIL) is a tumor-necrosis factor (TNF) family member and is a novel cytokine crucial in sustaining lymphocytic leukemia B cell survival and proliferation. However, its role in gastric cancer (GC) remains unclear. In this study, we investigated the expression pattern and prognostic role of APRIL in GC. METHODS: Expression of APRIL was assessed by immunohistochemistry and real-time PCR. Prognostic role of APRIL expression was evaluated. We also discovered the effect of APRIL on chemo-resistance in GC cells and the underlying mechanisms. RESULTS: APRIL mRNA levels were significantly increased in GC tissues compared with adjacent tissues and high expression levels of APRIL in tumor cells significantly correlated with poor overall survival in patients receiving cisplatin adjuvant treatment. Overexpression of APRIL in AGS cells significantly attenuated the therapeutic efficacy of cisplatin in vitro and in vivo. In contrast, silence of APRIL in SGC7901 cells enhanced cisplatin-induced tumor suppression. Our data further revealed that the canonical NF-κB pathway was involved in APRIL-mediated chemo-resistance. In addition, expression of APRIL was regulated by miR-145 in GC cells. CONCLUSION: APRIL is a novel clinical chemo-resistance biomarker for gastric cancer and might be a promising therapeutic target for GC patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Adult , Aged , Cell Line, Tumor , Female , Gene Silencing , Humans , Male , Middle Aged , NF-kappa B/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Treatment Outcome , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
13.
J Transl Med ; 12: 309, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25367394

ABSTRACT

BACKGROUND: MUC4 plays important roles in the malignant progression of human pancreatic cancer. But the huge length of MUC4 gene fragment restricts its functional and mechanism research. As one of its splice variants, MUC4/Y with coding sequence is most similar to that of the full-length MUC4 (FL-MUC4), together with alternative splicing of the MUC4 transcript has been observed in pancreatic carcinomas but not in normal pancreas. So we speculated that MUC4/Y might be involved in malignant progression similarly to FL-MUC4, and as a research model of MUC4 in pancreatic cancer. The conjecture was confirmed in the present study. METHODS: MUC4/Y expression was detected by real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) using gene-specific probe in the clinic samples. The effects of MUC4/Y were observed by serial in vitro and in vivo experiments based on stable over-expressed cell model. The underlying mechanisms were investigated by sequence-based transcriptome analysis and verified by qRT-PCR, Western blot and enzyme-linked immunosorbent assays. RESULTS: The detection of clinical samples indicates that MUC4/Y is significantly positive-correlated with tumor invasion and distant metastases. Based on stable forced-expressed pancreatic cancer PANC-1 cell model, functional studies show that MUC4/Y enhances malignant activity in vitro and in vivo, including proliferation under low-nutritional-pressure, resistance to apoptosis, motility, invasiveness, angiogenesis, and distant metastasis. Mechanism studies indicate the novel finding that MUC4/Y triggers malignancy-related positive feedback loops for concomitantly up-regulating the expression of survival factors to resist adverse microenvironment and increasing the expression of an array of cytokines and adhesion molecules to affect the tumor milieu. CONCLUSIONS: In light of the enormity of the potential regulatory circuitry in cancer afforded by MUC4 and/or MUC4/Y, repressing MUC4 transcription, inhibiting post-transcriptional regulation, including alternative splicing, or blocking various pathways simultaneously may be helpful for controlling malignant progression. MUC4/Y- expression model is proven to a valuable tool for the further dissection of MUC4-mediated functions and mechanisms.


Subject(s)
Mucin-4/genetics , Pancreatic Neoplasms/pathology , RNA Splicing , Signal Transduction , Transcriptome , Disease Progression , Feedback , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , RNA, Messenger/genetics
14.
Mol Cancer ; 13: 130, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24884523

ABSTRACT

BACKGROUND: Increasing evidence indicates an important role of transcription factor Yin Yang-1 (YY1) in human tumorigenesis. However, its function in cancer remains controversial and the relevance of YY1 to pancreatic ductal adenocarcinoma (PDAC) remains to be clarified. METHODS: In this study, we detected YY1 expression in clinical PDAC tissue samples and cell lines using quantitative RT-PCR, immunohistochemistry and western blotting. We also detected MUC4 and MMP10 mRNA levels in 108 PDAC samples using qRT-PCR and analyzed the correlations between YY1 and MUC4 or MMP10 expression. The role of YY1 in the proliferation, invasion and metastatic abilities of PDAC cells in vitro was studied by CCK-8 assay, cell migration and invasion assays. In vivo pancreatic tumor growth and metastasis was studied by a xenogenous subcutaneously implant model and a tail vein metastasis model. The potential mechanisms underlying YY1 mediated tumor progression in PDAC were explored by digital gene expression (DGE) sequencing, signal transduction pathways blockage experiments and luciferase assays. Statistical analysis was performed using the SPSS 15.0 software. RESULTS: We found that the expression of YY1 in PDACs was higher compared with their adjacent non-tumorous tissues and normal pancreas tissues. However, PDAC patients with high level overexpression of YY1 had better outcome than those with low level overexpression. YY1 expression levels were statistically negatively correlated with MMP10 expression levels, but not correlated with MUC4 expression levels. YY1 overexpression suppressed, whereas YY1 knockdown enhanced, the proliferation, invasion and metastatic properties of BXPC-3 cells, both in vitro and in vivo. YY1 suppresses invasion and metastasis of pancreatic cancer cells by downregulating MMP10 in a MUC4/ErbB2/p38/MEF2C-dependent mechanism. CONCLUSIONS: The present study suggested that YY1 plays a negative role, i.e. is a tumor suppressor, in PDAC, and may become a valuable diagnostic and prognostic marker of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Lung Neoplasms/genetics , Matrix Metalloproteinase 10/genetics , Pancreatic Neoplasms/genetics , YY1 Transcription Factor/genetics , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/secondary , Female , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/secondary , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Male , Matrix Metalloproteinase 10/metabolism , Mice , Mice, Nude , Middle Aged , Mucin-4/genetics , Mucin-4/metabolism , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction , Survival Analysis , Xenograft Model Antitumor Assays , YY1 Transcription Factor/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Oncol Rep ; 31(5): 2187-94, 2014 May.
Article in English | MEDLINE | ID: mdl-24676527

ABSTRACT

MUC4/Y, the transcript variant 4 of MUC4, lacks exon 2 as compared with the transcript variant 1 of MUC4. To date, direct evidence for the function of MU4/Y remains to be reported. Previous studies based their hypotheses regarding the function of MUC4/Y on the characteristic structure domains of this variant. The aim of the present study was to investigate the specific function of MUC4/Y. The pancreatic cancer cell line MIA PaCa-2 with low MUC4/Y expression was used to establish a stable cell model of MUC4/Y upregulation using a lentivirus vector system. Results showed that MUC4/Y anchored on the cytomembrane and affected cell morphology and cell cycle. Functional analyses indicated that MUC4/Y upregulation slightly potentiated cell proliferation and significantly suppressed apoptosis both in vivo and in vitro. Further studies revealed that the JNK and AKT signalling pathways were activated. Meanwhile, MUC4/Y upregulation elicited minimal effect on the phosphorylation level of HER2, a membrane partner of MUC4. These results suggest that MUC4/Y promotes tumour progression through its anti-apoptotic and weak mitogenic effect on MIA PaCa-2 cells.


Subject(s)
Apoptosis/genetics , Cell Proliferation/genetics , Mucin-4/genetics , Pancreatic Neoplasms/genetics , Animals , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Mucin-4/biosynthesis , Pancreatic Neoplasms/pathology , Phosphorylation/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Sequence Deletion/genetics , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
16.
FEBS Lett ; 588(5): 757-63, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24462679

ABSTRACT

Intestinal ischemic injury is a significant clinical problem arising from diseases or as a complication of abdominal surgery. Our previous study showed aquaporin 3 is involved in intestinal barrier impairment. Here, we revealed that intestinal ischemia induced a time-dependent increase of miR-874 expression and a time-dependent decrease of AQP3 expression, and the level of miR-874 expression was inversely related to AQP3 protein expression. In addition, miR-874 promoted the paracellular permeability in vitro through targeting 3'UTR of AQP3. Two of the tight junction proteins, Occludin and Claudin-1, were found to be involved in miR-874-induced intestinal barrier dysfunction.


Subject(s)
Aquaporin 3/genetics , Intestinal Mucosa/metabolism , Ischemia/metabolism , Mesenteric Vascular Occlusion/metabolism , MicroRNAs/physiology , Tight Junctions/metabolism , 3' Untranslated Regions , Animals , Aquaporin 3/metabolism , Bacterial Translocation , Base Sequence , Caco-2 Cells , Cell Hypoxia , Claudin-1/genetics , Claudin-1/metabolism , Gene Expression , Humans , Intestines/blood supply , Intestines/pathology , Mesenteric Arteries/pathology , Mice , Mice, Inbred C57BL , Occludin/genetics , Occludin/metabolism , RNA Interference
17.
Tumour Biol ; 35(5): 4389-99, 2014 May.
Article in English | MEDLINE | ID: mdl-24390615

ABSTRACT

Centrosomal protein 55 (CEP55) is the latest found member in the centrosomal relative protein family, which participates in cell-cycle regulation. CEP55 exists in many kinds of normal tissues and tumour cells such as hepatocellular carcinoma, and is important in carcinogenesis. However, the role of CEP55 in the pathogenesis of gastric cancer (GC) remains unclear. The mRNA levels of CEP55 in GC tissues and GC cell lines were examined by quantitative real-time PCR, and the protein expression of CEP55 in GC tissues was detected by Western blot and immunohistochemistry. The role of CEP55 in regulating the proliferation of GC cell lines was investigated both in vitro and in vivo. CEP55 was strongly upregulated in human GC, indicating that CEP55 contributed to carcinogenesis and progression of GC. Ectopic overexpression of CEP55 enhanced the cell proliferation, colony formation, and tumourigenicity of GC cells, whereas CEP55 knockdown inhibited these effects. We discovered that cell transformation induced by CEP55 was mediated by the AKT signalling pathway. Overexpression of CEP55 enhanced the phosphorylation of AKT and inhibited the activity of p21 WAF1/Cip1. In addition, cellular proliferation was suppressed as a result of cell cycle arrest at the G2/M phase in CEP55-knockdown cells. CEP55 expression was elevated in GC compared with normal control tissues. Credible evidence showed that CEP55 can be a potential therapeutic target in GC.


Subject(s)
Cell Cycle Proteins/physiology , Cell Proliferation , Nuclear Proteins/physiology , Stomach Neoplasms/pathology , Adult , Aged , Animals , Cell Cycle Checkpoints , Cell Cycle Proteins/analysis , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/physiology , Female , Humans , Male , Mice , Middle Aged , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction , Stomach Neoplasms/etiology
18.
Cancer Lett ; 346(1): 104-13, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24374017

ABSTRACT

The membrane mucin MUC4 is aberrantly expressed in multiple cancers and is of clinical significance to diagnosis and prognosis in pancreatic cancer. However, the role of MUC4 in angiogenesis and the potential association among these malignant capabilities have not been explored. In this study, we investigated the collective signaling mechanisms associated with MUC4-induced growth, metastasis and angiogenesis in pancreatic cancer. Knockdown of MUC4 in two pancreatic cancer cell lines led to downregulation of lysosomal degradation of E-cadherin by Src kinase through downregulation of pFAK and pSrc pathway. The downregulation of lysosomal degradation of E-cadherin in turn induced the formation of E-cadherin/ß-catenin complex and membrane translocation of ß-catenin, resulting in the downregulation of Wnt/ß-catenin signaling pathway. Thus, the Wnt/ß-catenin target genes c-Myc, Cyclin D1, CD44 and VEGF were down-regulated and their malignant functions proliferation, metastasis and angiogenesis were reduced. Taken together, MUC4-induced nuclear translocation of ß-catenin is a novel mechanism for growth, metastasis and angiogenesis of pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Mucin-4/metabolism , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/metabolism , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Heterografts , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Neoplasm Invasiveness/pathology , Protein Transport/physiology , RNA Interference , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology
19.
Mol Biol Rep ; 40(8): 4913-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23813057

ABSTRACT

The human mucin 4 (MUC4) is aberrantly expressed in pancreatic adenocarcinoma and tumor cell lines, while remaining undetectable in normal pancreas, indicating its important role in pancreatic cancer development. Although its transcriptional regulation has been investigated in considerable detail, some important elements remain unknown. The aim of the present study was to demonstrate the existence of a novel inhibitory element in the MUC4 promoter and characterize some of its binding proteins. By luciferase reporter assay, we located the inhibitory element between nucleotides -2530 and -2521 in the MUC4 promoter using a series of deletion and mutant reporter constructs. Electrophoretic mobility shift assay (EMSA) with Bxpc-3 cell nuclear extracts revealed that one protein or protein complex bind to this element. The proteins binding to this element were purified and identified as Yin Yang 1 (YY1) by mass spectrometry. Supershift assay and chromatin immunoprecipitation (ChIP) assay confirmed that YY1 binds to this element in vitro and in vivo. Moreover, transient YY1 overexpression significantly inhibited MUC4 promoter activity and endogenous MUC4 protein expression. In conclusion, we reported here a novel inhibitory element in the human MUC4 promoter. This provides additional data on MUC4 gene regulation and indicates that YY1 may be a potential target for abnormal MUC4 expression.


Subject(s)
Gene Expression Regulation/genetics , Mucin-4/metabolism , Promoter Regions, Genetic/genetics , YY1 Transcription Factor/metabolism , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Humans , Luciferases , Mass Spectrometry , Mucin-4/genetics , Oligonucleotides/genetics , Plasmids/genetics
20.
Cancer Epidemiol ; 34(4): 453-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20430715

ABSTRACT

OBJECTIVE: The objective is to apply synchrotron-based FTIR microspectroscopy and imaging to human hair tissue and investigate the possibility of the method in gastric cancer research and diagnosis. METHODS: Human hair from gastric cancer patients' scalp and normal persons' scalp were studied by synchrotron-based FTIR microspectroscopy and imaging. RESULTS: The micro-spectra and imaging show the difference between the normal and malignant hair tissues. Obvious peak shift of symmetric phosphate band is observed in micro-spectra of medulla region for the hair tissue of gastric cancer patients. Chemical imaging shows the distributions of lipid and amide II/v(s)PO(2)(-) have changed in the gastric cancer cases. CONCLUSIONS: The study indicates that the hair tissue's infrared microspectroscopy and imaging using synchrotron will be a potentially useful method for rapid early gastric cancer diagnosis.


Subject(s)
Diagnostic Imaging/instrumentation , Hair/chemistry , Lipids/analysis , Spectroscopy, Fourier Transform Infrared , Stomach Neoplasms/diagnosis , Synchrotrons , Amides/analysis , Case-Control Studies , Female , Humans , Male , Prognosis , Scalp/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...