Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Mol Carcinog ; 62(10): 1585-1598, 2023 10.
Article in English | MEDLINE | ID: mdl-37378449

ABSTRACT

Colorectal cancer is one of the most common malignancies worldwide. Liver metastasis is the major direct cause of colorectal cancer-related deaths. Although radical resection is the most effective treatment for colorectal cancer liver metastasis, several patients are not eligible for surgery. Therefore, there is a need to develop novel treatments based on the understanding of the biological mechanisms underlying liver metastasis in colorectal cancer. This study demonstrated that activin A/ACVR2A inhibits colon cancer cell migration and invasion, as well as suppresses the epithelial-to-mesenchymal transition of mouse colon cancer cells. This finding has been further validated in animal experiments. Mechanistic studies revealed that activin A binds to Smad2 (instead of Smad3) and activates its transcription. Analysis of the paired clinical samples further confirmed that the expression levels of ACVR2A and SMAD2 were the highest in adjacent healthy tissues, followed by primary colon cancer tissues and liver metastasis tissues, suggesting that ACVR2A downregulation may promote colon cancer metastasis. Bioinformatics analysis and clinical studies demonstrated that ACVR2A downregulation was significantly associated with liver metastasis and poor disease-free and progression-free survival of patients with colon cancer. These results suggest that the activin A/ACVR2A axis promotes colon cancer metastasis by selectively activating SMAD2. Thus, targeting ACVR2A is a potential novel therapeutic strategy to prevent colon cancer metastasis.


Subject(s)
Colonic Neoplasms , Liver Neoplasms , Animals , Mice , Activins/genetics , Activins/metabolism , Cell Line, Tumor , Cell Movement , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Liver Neoplasms/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Humans
2.
Cancer Lett ; 558: 216095, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36796670

ABSTRACT

Our previous research defined a novel metabolic cancer associated fibroblasts subset (meCAFs) enriched in loose-type pancreatic ductal adenocarcinoma (PDAC) and related to CD8+ T cells accumulation. Consistently, the abundance of meCAFs was associated with poor prognosis but better immunotherapy responses in PDAC patients. However, the metabolic characteristic of meCAFs and its cross-talk with CD8+ T cells remain to be elucidated. In this study, we identified PLA2G2A as a marker of meCAFs. In particular, the abundance of PLA2G2A+ meCAFs was positively related to the accumulation of total CD8+ T cells and negatively correlated with clinical outcomes of PDAC patients and infiltration of intratumoral CD8+ T cells. We demonstrated that PLA2G2A+ meCAFs substantially attenuated the antitumor ability of tumor infiltrating CD8+ T cells and facilitated tumor immune escape in PDAC. Mechanistically, PLA2G2A regulated the function of CD8+ T cells as a pivotal soluble mediator via MAPK/Erk and NF-κB signaling pathways. In conclusion, our study identified the unrecognized role of PLA2G2A+ meCAFs in promoting tumor immune escape by impeding the antitumor immune function of CD8+ T cells, and strongly suggested PLA2G2A as a promising biomarker and therapeutic target for immunotherapy in PDAC.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , T-Lymphocytes, Cytotoxic/metabolism , Cancer-Associated Fibroblasts/metabolism , CD8-Positive T-Lymphocytes , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Immunity , Tumor Microenvironment , Group II Phospholipases A2 , Pancreatic Neoplasms
3.
Biol Direct ; 17(1): 34, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36434686

ABSTRACT

BACKGROUND: The upregulated expression of CXCL1 has been validated in colorectal cancer patients. As a potential biotherapeutic target for colorectal cancer, the mechanism by which CXCL1 affects the development of colorectal cancer is not clear. METHODS: Expression data of CXCL1 in colorectal cancer were obtained from the GEO database and verified using the GEPIA database and the TIMER 2.0 database. Knockout and overexpression of CXCL1 in colorectal cancer cells by CRISPR/Cas and "Sleeping Beauty" transposon-mediated gene editing techniques. Cell biological function was demonstrated by CCK-8, transwell chamber and Colony formation assay. RT-qPCR and Western Blot assays measured RNA and protein expression. Protein localization and expression were measured by immunohistochemistry and immunofluorescence. RESULTS: Bioinformatics analysis showed significant overexpression of CXCL1 in the colorectal cancer tissues compared to normal human tissues, and identified CXCL1 as a potential therapeutic target for colorectal cancer. We demonstrate that CXCL1 promotes the proliferation and migration of colon cancer cells and has a facilitative effect on tumor angiogenesis. Furthermore, CXCL1 elevation promoted the migration of M2-tumor associated macrophages (TAMs) while disrupting the aggregation of CD4+ and CD8+ T cells at tumor sites. Mechanistic studies suggested that CXCL1 activates the NF-κB pathway. In the in vivo colon cancer transplantation tumor model, treatment with the P300 inhibitor C646 significantly inhibited the growth of CXCL1-overexpressing colon cancer. CONCLUSION: CXCL1 promotes colon cancer development through activation of NF-κB/P300, and that CXCL1-based therapy is a potential novel strategy to prevent colon cancer development.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Humans , NF-kappa B/metabolism , NF-kappa B/pharmacology , Signal Transduction , Colonic Neoplasms/genetics , Colorectal Neoplasms/metabolism , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Chemokine CXCL1/pharmacology
4.
Biochem Pharmacol ; 205: 115253, 2022 11.
Article in English | MEDLINE | ID: mdl-36176239

ABSTRACT

Lithocholic acid (LCA), one of the most common metabolic products of bile acids (BAs), is originally synthesized in the liver, stored in the gallbladder, and released to the intestine, where it assists absorption of lipid-soluble nutrients. LCA has recently emerged as a powerful reagent to inhibit tumorigenesis; however, the anti-tumor activity and molecular mechanisms of LCA in gallbladder cancer (GBC) remain poorly acknowledged. Here, we analyzed serum levels of LCA in human GBC and found that LCA was significantly downregulated in these patients, and reduced LCA levels were associated with poor clinical outcomes. Treatment of xenografts with LCA impeded tumor growth. Furthermore, LCA treatment in GBC cell lines decreased glutaminase (GLS) expression, glutamine (Gln) consumption, and GSH/GSSG and NADPH/NADP+ ratios, leading to cellular ferroptosis. In contrast, GLS overexpression in tumor cells fully restored GBC proliferation and decreased ROS imbalance, thus suppressing ferroptosis. Our findings reveal that LCA functions as a tumor-suppressive factor in GBC by downregulating GLS-mediated glutamine metabolism and subsequently inducing ferroptosis. This study may offer a new therapeutic strategy tailored to improve the treatment of GBC.


Subject(s)
Gallbladder Neoplasms , Glutaminase , Humans , Gallbladder Neoplasms/metabolism , Lithocholic Acid/pharmacology , Glutamine/metabolism , NADP , Reactive Oxygen Species , Glutathione Disulfide , Bile Acids and Salts , Cell Proliferation , Lipids , Cell Line, Tumor
5.
Front Genet ; 13: 882307, 2022.
Article in English | MEDLINE | ID: mdl-35783274

ABSTRACT

Background: Mining the prognostic biomarkers of colorectal cancer (CRC) has important clinical and scientific significance. The role of Fc receptor-like B (FCRLB) in solid tumors has never been reported or studied to our knowledge, and the prognostic role of FCRLB in CRC still awaits characterization. Methods: The potential prognostic factor FCRLB was screened out through TCGA database analysis. Then, its expression and associations with clinicopathological variables were assessed in the TCGA CRC cohort. The prognostic value of FCRLB was examined with multiple methods, such as the Kaplan-Meier method, ROC curve, time-dependent ROC analysis, and prediction model nomograms. Then, functional enrichment and annotation among the high and low FCRLB groups were achieved utilizing GO and KEGG analyses and GSEA. Fresh CRC tissue samples obtained clinically were used for the preparation of the tissue microarray and for further validation. Results: FCRLB was highly expressed in CRC tissues compared to normal tissues. Moreover, over-expression of FCRLB correlated with higher CEA levels, advanced T stage, N stage, M stage, AJCC stage, lymphatic invasion, perineural invasion, and incomplete resection (R1 and R2 resection). In addition, high expression of FCRLB was closely correlated to less favorable OS, DSS, and PFI. The analysis of CRC tissue microarray further confirmed the conclusion drawn from the TCGA data analysis. Conclusion: FCRLB is notably up-regulated in CRC tissues and may serve as a potential biomarker of CRC.

7.
Sensors (Basel) ; 22(11)2022 May 28.
Article in English | MEDLINE | ID: mdl-35684740

ABSTRACT

Reference-based image super-resolution (RefSR) methods have achieved performance superior to that of single image super-resolution (SISR) methods by transferring texture details from an additional high-resolution (HR) reference image to the low-resolution (LR) image. However, existing RefSR methods simply add or concatenate the transferred texture feature with the LR features, which cannot effectively fuse the information of these two independently extracted features. Therefore, this paper proposes a dual projection fusion for reference-based image super-resolution (DPFSR), which enables the network to focus more on the different information between feature sources through inter-residual projection operations, ensuring effective filling of detailed information in the LR feature. Moreover, this paper also proposes a novel backbone called the deep channel attention connection network (DCACN), which is capable of extracting valuable high-frequency components from the LR space to further facilitate the effectiveness of image reconstruction. Experimental results show that we achieve the best peak signal-to-noise ratio (PSNR) and structure similarity (SSIM) performance compared with the state-of-the-art (SOTA) SISR and RefSR methods. Visual results demonstrate that the proposed method in this paper recovers more natural and realistic texture details.

8.
Foods ; 11(9)2022 May 03.
Article in English | MEDLINE | ID: mdl-35564052

ABSTRACT

"Fo Tiao Qiang" is a famous dish with Chinese characteristics. It is delicious, rich in materials, and high in nutritional value. Through physical and chemical analysis, electronic tongue, gas chromatography-ion mobility spectroscopy, and other technologies, the present study explored the quality characteristics and flavor differences of Fo Tiao Qiang by using different thawing methods (natural thawing, ultrasonic thawing, microwave thawing, and water bath thawing). The results show that the protein content was slightly higher in Fo Tiao Qiang with ultrasonic thawing than others. The fat content of the microwave-thawed Fo Tiao Qiang was significantly lower than the other three kinds of samples. After ultrasonic thawing, the number of free amino acids in the samples were the highest and the umami taste was the best. Compared with natural thawing, most of the flavor substances decreased in ultrasonic thawing, microwave thawing, and water bath thawing. However, several substances increased, such as alpha-terpineol, beta-phenylethyl alcohol, phenylacetaldehyde, cis-rose oxide, isobutyl acetate, and 2-3-pentanedione. This study revealed the changing laws of different thawing methods on the quality characteristics and flavor characteristics of Fo Tiao Qiang. It provides theoretical guidance for the industrial production and quality control of Fo Tiao Qiang.

9.
J Exp Clin Cancer Res ; 40(1): 373, 2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34823564

ABSTRACT

BACKGROUND: Gallbladder cancer (GBC) is known for its high malignancy and multidrug resistance. Previously, we uncovered that impaired integrity and stability of the elongator complex leads to GBC chemotherapy resistance, but whether its restoration can be an efficient therapeutic strategy for GBC remains unknown. METHODS: RT-qPCR, MS-qPCR and ChIP-qPCR were used to evaluate the direct association between ELP5 transcription and DNA methylation in tumour and non-tumour tissues of GBC. EMSA, chromatin accessibility assays, and luciferase assays were utilized to analysis the DNA methylation in interfering PAX5-DNA interactions. The functional experiments in vitro and in vivo were performed to investigate the effects of DNA demethylating agent decitabine (DAC) on the transcription activation of elongator complex and the enhanced sensitivity of gemcitabine in GBC cells. Tissue microarray contains GBC tumour tissues was used to evaluate the association between the expression of ELP5, DNMT3A and PAX5. RESULTS: We demonstrated that transcriptional repression of ELP5 in GBC was highly correlated with hypermethylation of the promoter. Mechanistically, epigenetic analysis revealed that DNA methyltransferase DNMT3A-catalysed hypermethylation blocked transcription factor PAX5 activation of ELP5 by disrupting PAX5-DNA interaction, resulting in repressed ELP5 transcription. Pharmacologically, the DNA demethylating agent DAC eliminated the hypermethylated CpG dinucleotides in the ELP5 promoter and then facilitated PAX5 binding and reactivated ELP5 transcription, leading to the enhanced function of the elongator complex. To target this mechanism, we employed a sequential combination therapy of DAC and gemcitabine to sensitize GBC cells to gemcitabine-therapy through epigenetic activation of the elongator complex. CONCLUSIONS: Our findings suggest that ELP5 expression in GBC is controlled by DNA methylation-sensitive induction of PAX5. The sequential combination therapy of DAC and gemcitabine could be an efficient therapeutic strategy to overcome chemotherapy resistance in GBC.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Epigenomics/methods , Gallbladder Neoplasms/drug therapy , Animals , Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Gallbladder Neoplasms/genetics , Humans , Male , Mice , Mice, Nude , Gemcitabine
10.
Front Immunol ; 11: 1087, 2020.
Article in English | MEDLINE | ID: mdl-32636834

ABSTRACT

Regulatory T cells (Treg cells) are essential for maintaining immune tolerance, and the dysfunction of Treg cells may cause autoimmune diseases and tumors. Forkhead box P3 (FOXP3) is the key transcription factor controlling Treg cell development and suppressive function. Mouse double minute 2 homolog (MDM2), an E3 ubiquitin ligase, has been identified as an oncoprotein that mediates the ubiquitination and degradation of tumor suppressor p53; however, whether it has functions in Treg cells remains unknown. Here, we demonstrate that MDM2 positively regulates human Treg cell suppressive function via its mediated ubiquitination and stabilization of FOXP3. Knockdown of MDM2 with shRNA in human primary Treg cells leads to the impaired ability of FOXP3 to regulate the expression levels of downstream genes and the attenuated suppressive capacity of Treg cells, due to FOXP3 instability. Consistently, MDM2 overexpression in human Treg cells enhances FOXP3 stability and Treg cell suppressive capacity. Mechanistically, MDM2 interacts with FOXP3, and mainly mediates monoubiquitination and polyubiquitination of FOXP3, thus stabilizing the protein level of FOXP3. We have also found lysine residues in FOXP3 required for MDM2-mediated ubiquitination. In addition, TCR/CD28 signaling upregulates the expression level of MDM2 and its mediated FOXP3 ubiquitination in human Treg cells. Therefore, our findings reveal that MDM2 in Treg cells could be a potential therapeutic target for treating autoimmune diseases and tumors.


Subject(s)
Forkhead Transcription Factors/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Amino Acid Sequence , CD28 Antigens/metabolism , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Immune Tolerance , In Vitro Techniques , Lysine/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Stability , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Ubiquitination
11.
Oncogene ; 39(26): 4983-5000, 2020 06.
Article in English | MEDLINE | ID: mdl-32514152

ABSTRACT

Bile acids (BAs), well-defined signaling molecules with diverse metabolic functions, play important roles in cellular processes associated with many cancers. As one of the most common BAs, deoxycholic acid (DCA) is originally synthesized in the liver, stored in the gallbladder, and processed in the gut. DCA plays crucial roles in various tumors; however, functions and molecular mechanisms of DCA in gallbladder cancer (GBC) still remain poorly characterized. Here, we analyzed human GBC samples and found that DCA was significantly downregulated in GBC, and reduced levels of DCA was associated with poor clinical outcome in patients with GBC. DCA treatment impeded tumor progression by halting cell proliferation. DCA decreased miR-92b-3p expression in an m6A-dependent posttranscriptional modification manner by facilitating dissociation of METTL3 from METTL3-METTL14-WTAP complex, which increased the protein level of the phosphatase and tensin homolog, a newly identified target of miR-92b-3p, and subsequently inactivated the PI3K/AKT signaling pathway. Our findings revealed that DCA might function as a tumor suppressive factor in GBC at least by interfering with miR-92b-3p maturation, and suggested that DCA treatment could provide a new therapeutic strategy for GBC.


Subject(s)
Adenosine/analogs & derivatives , Cell Proliferation/drug effects , Deoxycholic Acid/pharmacology , Gallbladder Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , MicroRNAs/genetics , Adenosine/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Deoxycholic Acid/metabolism , Disease Progression , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/therapy , HEK293 Cells , Humans , Male , Mice, Inbred BALB C , Mice, Nude , RNA Interference , Xenograft Model Antitumor Assays/methods
12.
J Cell Mol Med ; 24(2): 1599-1613, 2020 01.
Article in English | MEDLINE | ID: mdl-31782270

ABSTRACT

Gallbladder cancer (GBC) is the leading malignancy of biliary system showing refractory chemoresistance to current first-line drugs. Growing epidemiological evidences have established that the incidence of GBC exhibits significant gender predominance with females two-threefold higher than males, suggesting oestrogen/oestrogen receptors (ERs) signalling might be a critical driver of tumorigenesis in gallbladder. This study aims to evaluate the antitumour activity of tamoxifen (TAM), a major agent of hormonal therapy for breast cancer, in preclinical GBC model. Quantitative real-time PCR was used to investigate mRNA levels. Protein expression was measured by immunohistochemistry and Western blot. Glycolytic levels were measured by glucose consumption and lactic acid measurement. The antitumour activity of TAM alone or with cisplatin was examined with CCK8 assay, colony formation, flow cytometry and in vivo models. The results revealed that ERɑ expression was higher in GBC tissues and predicted poor clinical outcomes. TAM was showed effective against a variety of GBC cell lines. Mechanical investigations revealed that TAM enabled potent reactive oxygen species (ROS) production by reduced nuclear factor Nrf2 expression and its target genes, leading to the activation of AMPK, which subsequently induced impaired glycolysis and survival advantages. Notably, TAM was demonstrated to sensitize GBC cells to cisplatin (CDDP) both in vitro and in vivo. In agreement with these findings, elimination of oestrogens by ovariectomy in nude mice prevented CDDP resistance. In summary, these results provide basis for TAM treatment for GBC and shed novel light on the potential application of endocrine therapy for patients with GBC.


Subject(s)
Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/pathology , Glucose/metabolism , Tamoxifen/pharmacology , Adenylate Kinase/metabolism , Aged , Apoptosis/drug effects , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Estrogen Receptor alpha/metabolism , Female , Gallbladder Neoplasms/enzymology , Glycolysis/drug effects , Humans , Male , Middle Aged , Models, Biological , Molecular Targeted Therapy , Multivariate Analysis , NF-E2-Related Factor 2/metabolism , Ovariectomy , Prognosis , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Burden , Xenograft Model Antitumor Assays
13.
Nat Commun ; 10(1): 5492, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31792210

ABSTRACT

Gemcitabine is the first-line treatment for locally advanced and metastatic gallbladder cancer (GBC), but poor gemcitabine response is universal. Here, we utilize a genome-wide CRISPR screen to identify that loss of ELP5 reduces the gemcitabine-induced apoptosis in GBC cells in a P53-dependent manner through the Elongator complex and other uridine 34 (U34) tRNA-modifying enzymes. Mechanistically, loss of ELP5 impairs the integrity and stability of the Elongator complex to abrogate wobble U34 tRNA modification, and directly impedes the wobble U34 modification-dependent translation of hnRNPQ mRNA, a validated P53 internal ribosomal entry site (IRES) trans-acting factor. Downregulated hnRNPQ is unable to drive P53 IRES-dependent translation, but rescuing a U34 modification-independent hnRNPQ mutant could restore P53 translation and gemcitabine sensitivity in ELP5-depleted GBC cells. GBC patients with lower ELP5, hnRNPQ, or P53 expression have poor survival outcomes after gemcitabine chemotherapy. These results indicate that the Elongator/hnRNPQ/P53 axis controls gemcitabine sensitivity in GBC cells.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Carrier Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Deoxycytidine/analogs & derivatives , Gallbladder Neoplasms/drug therapy , Gallbladder Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cohort Studies , Deoxycytidine/administration & dosage , Female , Gallbladder Neoplasms/metabolism , Genome-Wide Association Study , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Internal Ribosome Entry Sites , Male , Middle Aged , RNA Processing, Post-Transcriptional , RNA, Transfer/genetics , RNA, Transfer/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Gemcitabine
14.
Mol Cancer ; 18(1): 166, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31752867

ABSTRACT

BACKGROUND: CircRNAs are found to affect initiation and progression of several cancer types. However, whether circRNAs are implicated in gallbladder cancer (GBC) progression remains obscure. METHODS: We perform RNA sequencing in 10 pairs of GBC and para-cancer tissues. CCK8 and clone formation assays are used to evaluate proliferation ability of GBC cells. qPCR and Western blot are used to determine expression of RNAs and proteins, respectively. CircRNA-protein interaction is confirmed by RNA pulldown, RNA immunoprecipitation, and fluorescence in situ hybridization. RESULTS: We find that circRNA expression pattern is tremendously changed during GBC development. Among dozens of significantly changed circRNAs, a circRNA generated from the oncogene ERBB2, named as circERBB2, is one of the most significant changes. CircERBB2 promotes GBC proliferation, in vitro and in vivo. Other than being a miRNA sponge, circERBB2 accumulates in the nucleoli and regulates ribosomal DNA transcription, which is one of the rate-limiting steps of ribosome synthesis and cellular proliferation. CircERBB2 regulates nucleolar localization of PA2G4, thereby forming a circERBB2-PA2G4-TIFIA regulatory axis to modulate ribosomal DNA transcription and GBC proliferation. Increased expression of circERBB2 is associated with worse prognosis of GBC patients. CONCLUSIONS: Our findings demonstrate that circERBB2 serves as an important regulator of cancer cell proliferation and shows the potential to be a new therapeutic target of GBC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA, Ribosomal , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , RNA, Circular , RNA-Binding Proteins/metabolism , Receptor, ErbB-2/genetics , Alternative Splicing , Biomarkers, Tumor , Cell Line, Tumor , Disease Progression , Gallbladder Neoplasms/pathology , Gene Expression Profiling , Humans , Models, Biological , Prognosis , ROC Curve
15.
J Exp Clin Cancer Res ; 38(1): 247, 2019 Jun 10.
Article in English | MEDLINE | ID: mdl-31182136

ABSTRACT

BACKGROUND: Gallbladder cancer (GBC) is an extremely malignant tumor with a high mortality rate. Little is known about its invasion and metastasis mechanism so far. METHODS: To identify the driver genes in GBC metastasis, we performed a mRNA microarray of metastatic GBC and paired non-tumor samples, and found PLEK2 was markedly upregulated in GBC tissues. Next, the expression of PLEK2 in GBC were examined in a larger cohort of patients by qRT-PCR, western blot and IHC staining. The clinicopathologic correlation of PLEK2 was determined by statistical analyses. The biological involvement of PLEK2 in GBC metastasis and the underlying mechanisms were investigated. RESULTS: In this study, we found that PLEK2 had higher expression in GBC tumor tissues compared to non-cancerous adjacent tissues and cholecystolithiasis tissues. The clinicopathologic analyses showed PLEK2 expression was positively correlated with tumor TNM stage, distant metastasis and PLEK2 was an independent predictor of overall survival (OS) in GBC patients. The cellular function assays showed PLEK2 promoted GBC cells migration, invasion and liver metastasis in mouse model via the regulation of epithelial-mesenchymal transition (EMT) process. Our mass spectrum and co-immunoprecipitation (co-IP) assays demonstrated that PLEK2 could interact with the kinase domain of EGFR and suppress EGFR ubiquitination mediated by c-CBL, leading to constitutive activation of EGFR signaling. Furthermore, RNA-sequencing and qRT-PCR results demonstrated chemokine (C-C motif) ligand 2 (CCL2), a target gene downstream of PLEK2/EGFR signaling, mediated the motility-promoting function of PLEK2. CONCLUSIONS: On the basis of these collective data, we propose that PLEK2 promotes the invasion and metastasis of GBC by EGFR/CCL2 pathway and PLEK2 can serve as a potential therapeutic target for GBC treatment.


Subject(s)
Chemokine CCL2/metabolism , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/pathology , Membrane Proteins/metabolism , Signal Transduction , Aged , Aged, 80 and over , Animals , Biomarkers , Cell Line, Tumor , Cell Movement , Cell Proliferation , ErbB Receptors/metabolism , Gallbladder Neoplasms/mortality , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Protein Binding
16.
J Biol Chem ; 293(52): 20099-20111, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30389786

ABSTRACT

The CD4+CD25+FOXP3+ regulatory T (Treg) cells are critical for maintaining immune tolerance in healthy individuals and are reported to restrict anti-inflammatory responses and thereby promote tumor progression, suggesting them as a target in the development of antitumor immunotherapy. Forkhead box P3 (FOXP3) is a key transcription factor governing Treg lineage differentiation and their immune-suppressive function. Here, using Treg cells, as well as HEK-293T and Jurkat T cells, we report that the stability of FOXP3 is directly and positively regulated by the E3 ubiquitin ligase ring finger protein 31 (RNF31), which catalyzes the conjugation of atypical ubiquitin chains to the FOXP3 protein. We observed that shRNA-mediated RNF31 knockdown in human Treg cells decreases FOXP3 protein levels and increases levels of interferon-γ, resulting in a Th1 helper cell-like phenotype. Human Treg cells that ectopically expressed RNF31 displayed stronger immune-suppressive capacity, suggesting that RNF31 positively regulates both FOXP3 stability and Treg cell function. Moreover, we found that RNF31 is up-regulated in Treg cells that infiltrate human gastric tumor tissues compared with their counterparts residing in peripheral and normal tissue. We also found that elevated RNF31 expression in intratumoral Treg cells is associated with poor survival of gastric cancer patients, suggesting that RNF31 supports the immune-suppressive functions of Treg cells. Our results suggest that RNF31 could be a potential therapeutic target in immunity-based interventions against human gastric cancer.


Subject(s)
Forkhead Transcription Factors/immunology , Gene Expression Regulation, Enzymologic/immunology , T-Lymphocytes, Regulatory/immunology , Ubiquitin-Protein Ligases/immunology , Ubiquitination/immunology , Up-Regulation/immunology , Disease-Free Survival , HEK293 Cells , Humans , Jurkat Cells , Protein Stability , Stomach Neoplasms/immunology , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Rate , T-Lymphocytes, Regulatory/pathology
17.
Oncotarget ; 8(41): 71197-71205, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-29050356

ABSTRACT

Negative lymph node (NLN) count has been validated as a protective predictor in various cancers after radical resection. However, the prognostic value of NLN count in the setting of stage IV gastric cancer patients who have received palliative resection has not been investigated. Surveillance, Epidemiology, and End Results Program (SEER)-registered gastric cancer patients were used for analysis in this study. Kaplan-Meier survival curves and multivariate Cox proportional hazards model were used to assess the risk factors for patients' survivals. The results showed that NLN count and N stage were independently prognostic factors in patients with stage IV gastric cancer after palliative surgery (P< 0.001). X-tile plots identified 2 and 11 as the optimal cutoff values to divide the patients into high, middle and low risk subsets in term of cause-specific survival (CSS). And NLN count was proved to be an independently prognostic factor in multivariate Cox analysis (P< 0.001). The risk score of NLN counts demonstrated that the plot of hazard ratios (HRs) for NLN counts sharply increased when the number of NLN counts decreased. Collectively, our present study revealed that NLN count was an independent prognostic predictor in stage IV gastric cancer after palliative resection. Standard lymph node dissection, such as D2 lymphadectomy maybe still necessary during palliative resection for patients with metastatic gastric cancer.

18.
FEBS Lett ; 591(12): 1677-1686, 2017 06.
Article in English | MEDLINE | ID: mdl-28477415

ABSTRACT

CD4+ CD25+ regulatory T (Treg) cells comprise a unique subset of T cells required for maintaining immune homeostasis. However, the molecular mechanisms associated with the functional variety of Treg cells are not fully delineated. In the present study, we demonstrate that ubiquitin-specific protease (USP)4 physically interacted with interferon regulatory factor 8 (IRF8) function via a K48-linked deubiquitinase, which stabilized IRF8 protein levels in Treg cells. Depletion of USP4 promoted the polyubiquitination of IRF8 and the upregulation of type 2 inflammatory cytokine gene expression in Treg cells. Consistently, treatment of Treg cells with USP4 inhibitor facilitated the polyubiquitination of IRF8. In addition, the deficiency of USP4 alleviated the suppressive function of Treg cells. Taken together, our results suggest that USP4 interacts with and stabilizes IRF8 to promote the suppressive function of Treg cells.


Subject(s)
Interferon Regulatory Factors/metabolism , T-Lymphocytes, Regulatory/metabolism , Ubiquitin-Specific Proteases/metabolism , Amino Acid Substitution , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Interferon Regulatory Factors/chemistry , Interferon Regulatory Factors/genetics , Lysine/metabolism , Mutagenesis, Site-Directed , Mutation , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Stability/drug effects , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/chemistry , Ubiquitin-Specific Proteases/genetics , Ubiquitination/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...