Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
FASEB J ; 38(10): e23667, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38742812

ABSTRACT

Immunity imbalance of T helper 17 (Th17)/regulatory T (Treg) cells is involved in the pathogenesis of Crohn's disease (CD). Complanatuside A (CA), a flavonol glycoside, exerts anti-inflammatory activities and our study aimed to identify its effect on TNBS-induced colitis and the possible mechanisms. We found that CA alleviated the symptoms of colitis in TNBS mice, as demonstrated by prevented weight loss and colon length shortening, as well as decreased disease activity index scores, inflammatory scores, and levels of proinflammatory factors. Flow cytometry analysis showed that CA markedly reduced the percentage of Th17 cells while increasing the percentage of Treg cells in TNBS mice. Under Th17 cell polarizing conditions, CA inhibited the differentiation of Th17 cells while the Treg cell differentiation was elevated under Treg cell polarizing conditions. Furthermore, it was observed that JAK2 interacted with CA through six hydrogen bonds via molecular docking. The phosphorylation of JAK2/STAT3 was reduced by CA, which might be correlated with the protective effect of CA on colitis. In conclusion, CA reduced the imbalance of Th17/Treg cells by inhibiting the JAK2/STAT3 signaling pathway in TNBS-induced colitis, which may provide novel strategies for CD treatment.


Subject(s)
Colitis , Janus Kinase 2 , STAT3 Transcription Factor , Signal Transduction , T-Lymphocytes, Regulatory , Th17 Cells , Animals , Male , Mice , Cell Differentiation/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Janus Kinase 2/metabolism , Mice, Inbred BALB C , Signal Transduction/drug effects , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism , Trinitrobenzenesulfonic Acid
2.
FASEB J ; 38(14): e23817, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39003633

ABSTRACT

Excessive apoptosis of intestinal epithelial cells leads to intestinal barrier dysfunction, which is not only one of the pathological features of inflammatory bowel disease (IBD) but also a therapeutic target. A natural plant extract, Ginkgetin (GK), has been reported to have anti-apoptotic activity, but its role in IBD is unknown. This study aimed to explore whether GK has anti-colitis effects and related mechanisms. An experimental colitis model induced by dextran sulfate sodium (DSS) was established, and GK was found to relieve colitis in DSS-induced mice as evidenced by improvements in weight loss, colon shortening, Disease Activity Index (DAI), macroscopic and tissue scores, and proinflammatory mediators. In addition, in DSS mice and TNF-α-induced colonic organoids, GK protected the intestinal barrier and inhibited intestinal epithelial cell apoptosis, by improving permeability and inhibiting the number of apoptotic cells and the expression of key apoptotic regulators (cleaved caspase 3, Bax and Bcl-2). The underlying mechanism of GK's protective effect was explored by bioinformatics, rescue experiments and molecular docking, and it was found that GK might directly target and activate EGFR, thereby interfering with PI3K/AKT signaling to inhibit apoptosis of intestinal epithelial cells in vivo and in vitro. In conclusion, GK inhibited intestinal epithelial apoptosis in mice with experimental colitis, at least in part, by activating EGFR and interfering with PI3K/AKT activation, explaining the underlying mechanism for ameliorating colitis, which may provide new options for the treatment of IBD.


Subject(s)
Apoptosis , Biflavonoids , Colitis , Dextran Sulfate , Epithelial Cells , ErbB Receptors , Intestinal Mucosa , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Apoptosis/drug effects , Mice , Proto-Oncogene Proteins c-akt/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis/pathology , ErbB Receptors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Dextran Sulfate/toxicity , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Biflavonoids/pharmacology , Biflavonoids/therapeutic use , Male , Humans
3.
FASEB J ; 37(6): e22948, 2023 06.
Article in English | MEDLINE | ID: mdl-37130016

ABSTRACT

Bryostatin-1 (Bryo-1) exerts antioxidative stress effects in multiple diseases, and we confirmed that it improves intestinal barrier dysfunction in experimental colitis. Nevertheless, there are few reports on its action on intestinal ischemia/reperfusion (I/R). In this study, we mainly explored the effect of Bryo-1 on intestinal I/R injury and determined the mechanism. C57BL/6J mice underwent temporary superior mesenteric artery (SMA) obturation to induce I/R, on the contrary, Caco-2 cells suffered to oxygen and glucose deprivation/reperfusion (OGD/R) to establish the in vitro model. RAW264.7 cells were stimulated with LPS to induce macrophage inflammation. The drug gradient experiment was used to demonstrate in vivo and in vitro models. Bryo-1 ameliorated the intestinal I/R-induced injury of multiple organs and epithelial cells. It also alleviated intestinal I/R-induced barrier disruption of intestines according to the histology, intestinal permeability, intestinal bacterial translocation rates, and tight junction protein expression results. Bryo-1 significantly inhibited oxidative stress damages and inflammation, which may contribute to the restoration of intestinal barrier function. Further, Bryo-1 significantly activated Nrf2/HO-1 signaling in vivo. However, the deletion of Nrf2 in Caco-2 and RAW264.7 cells attenuated the protective functions of Bryo-1 and significantly abolished the anti-inflammatory effect of Bryo-1 on LPS-induced macrophage inflammation. Bryo-1 protects intestines against I/R-induced injury. It is associated with intestinal barrier protection, as well as inhibition of inflammation and oxidative stress partly through Nrf2/HO-1 signaling.


Subject(s)
Intestinal Diseases , Reperfusion Injury , Animals , Humans , Mice , Bryostatins/pharmacology , Caco-2 Cells , Inflammation/metabolism , Intestinal Diseases/prevention & control , Ischemia , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Reperfusion , Reperfusion Injury/metabolism
4.
Radiol Med ; 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-38997568

ABSTRACT

BACKGROUND: The accurate identification of microvascular invasion (MVI) in patients with hepatocellular carcinoma (HCC) is of great clinical importance. PURPOSE: To develop a radiomics nomogram based on susceptibility-weighted imaging (SWI) and T2-weighted imaging (T2WI) for predicting MVI in early-stage (Barcelona Clinic Liver Cancer stages 0 and A) HCC patients. MATERIALS AND METHODS: A prospective cohort of 189 participants with HCC was included for model training and testing, and an additional 34 participants were enrolled for external validation. ITK-SNAP was used to manually segment the tumour, and PyRadiomics was used to extract radiomic features from the SWI and T2W images. Variance filtering, student's t test, least absolute shrinkage and selection operator regression and random forest (RF) were applied to select meaningful features. Four machine learning classifiers, including K-nearest neighbour, RF, logistic regression and support vector machine-based models, were established. Independent clinical and radiological risk factors were also determined to establish a clinical model. The best radiomics and clinical models were further evaluated in the validation set. In addition, a nomogram was constructed from the radiomic model and independent clinical factors. Diagnostic efficacy was evaluated by receiver operating characteristic curve analysis with fivefold cross-validation. RESULTS: AFP levels greater than 400 ng/mL [odds ratio (OR) 2.50; 95% confidence interval (CI) 1.239-5.047], tumour diameter greater than 5 cm (OR 2.39; 95% CI 1.178-4.839), and absence of pseudocapsule (OR 2.053; 95% CI 1.007-4.202) were found to be independent risk factors for MVI. The areas under the curve (AUCs) of the best radiomic model were 1.000 and 0.882 in the training and testing cohorts, respectively, while those of the clinical model were 0.688 and 0.6691. In the validation set, the radiomic model achieved better diagnostic performance (AUC = 0.888) than the clinical model (AUC = 0.602). The combination of clinical factors and the radiomic model yielded a nomogram with the best diagnostic performance (AUC = 0.948). CONCLUSION: SWI and T2WI-derived radiomic features are valuable for noninvasively and accurately identifying MVI in early-stage HCC. Furthermore, the integration of radiomics and clinical factors yielded a predictive nomogram with satisfactory diagnostic performance and potential clinical benefits.

5.
J Magn Reson Imaging ; 2023 Oct 27.
Article in English | MEDLINE | ID: mdl-37888871

ABSTRACT

BACKGROUND: The metastatic vascular patterns of hepatocellular carcinoma (HCC) are mainly microvascular invasion (MVI) and vessels encapsulating tumor clusters (VETC). However, most existing VETC-related radiological studies still focus on the prediction of VETC status. PURPOSE: This study aimed to build and compare VETC-MVI related models (clinical, radiomics, and deep learning) associated with recurrence-free survival of HCC patients. STUDY TYPE: Retrospective. POPULATION: 398 HCC patients (349 male, 49 female; median age 51.7 years, and age range: 22-80 years) who underwent resection from five hospitals in China. The patients were randomly divided into training cohort (n = 358) and test cohort (n = 40). FIELD STRENGTH/SEQUENCE: 3-T, pre-contrast T1-weighted imaging spoiled gradient recalled echo (T1WI SPGR), T2-weighted imaging fast spin echo (T2WI FSE), and contrast enhanced arterial phase (AP), delay phase (DP). ASSESSMENT: Two radiologists performed the segmentation of HCC on T1WI, T2WI, AP, and DP images, from which radiomic features were extracted. The RFS related clinical characteristics (VETC, MVI, Barcelona stage, tumor maximum diameter, and alpha fetoprotein) and radiomic features were used to build the clinical model, clinical-radiomic (CR) nomogram, deep learning model. The follow-up process was done 1 month after resection, and every 3 months subsequently. The RFS was defined as the date of resection to the date of recurrence confirmed by radiology or the last follow-up. Patients were followed up until December 31, 2022. STATISTICAL TESTS: Univariate COX regression, least absolute shrinkage and selection operator (LASSO), Kaplan-Meier curves, log-rank test, C-index, and area under the curve (AUC). P < 0.05 was considered statistically significant. RESULTS: The C-index of deep learning model achieved 0.830 in test cohort compared with CR nomogram (0.731), radiomic signature (0.707), and clinical model (0.702). The average RFS of the overall patients was 26.77 months (range 1-80 months). DATA CONCLUSION: MR deep learning model based on VETC and MVI provides a potential tool for survival assessment. EVIDENCE LEVEL: 3 TECHNICAL EFFICACY: Stage 3.

6.
FASEB J ; 36(11): e22589, 2022 11.
Article in English | MEDLINE | ID: mdl-36197455

ABSTRACT

Intestinal inflammation and intestinal barrier damage are important pathological changes in Crohn's disease (CD). Vindoline is a natural monomer with anti-inflammatory effects. We employed CD model mice to explore the effect of Vindoline on CD-like colitis and the possible mechanism. Il-10-deficient (Il-10-/- ) mice and wild-type (WT) mice (both aged 15 weeks, male) were used to explore the effect of Vindoline on colitis and intestinal barrier damage, as well as macrophage-mediated inflammation. Bone-marrow-derived macrophages (BMDMs) and colonic organoids from mice were used to explore the inhibitory effect of Vindoline on macrophage-mediated inflammation and the protective effect on inflammation-induced intestinal barrier damage as well as the possible mechanism. We found that Vindoline significantly ameliorated colitis in CD mice, as evidenced by increased weight change and colon length and decreased the colon macroscopic injury score, histological inflammatory score, and the expression of pro-inflammatory mediators. Vindoline also protected against intestinal barrier damage in CD mice. Furthermore, Vindoline inhibited macrophage-mediated inflammation and protected against inflammation-induced intestinal barrier damage in the coculture system. In addition, Vindoline ameliorated colitis in CD mice by protecting against inflammation-induced intestinal barrier damage, which may be caused by inhibition of MAPK signaling pathway. This protective effect suggests that Vindoline has potential value for clinical application in the treatment of CD.


Subject(s)
Colitis , Crohn Disease , Animals , Anti-Inflammatory Agents/pharmacology , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Colon/metabolism , Crohn Disease/drug therapy , Crohn Disease/pathology , Dextran Sulfate/pharmacology , Disease Models, Animal , Inflammation/pathology , Inflammation Mediators/pharmacology , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Vinblastine/analogs & derivatives
7.
Eur Radiol ; 33(7): 4949-4961, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36786905

ABSTRACT

OBJECTIVES: The accurate prediction of post-hepatectomy early recurrence in patients with hepatocellular carcinoma (HCC) is crucial for decision-making regarding postoperative adjuvant treatment and monitoring. We aimed to explore the feasibility of deep learning (DL) features derived from gadoxetate disodium (Gd-EOB-DTPA) MRI, qualitative features, and clinical variables for predicting early recurrence. METHODS: In this bicentric study, 285 patients with HCC who underwent Gd-EOB-DTPA MRI before resection were divided into training (n = 195) and validation (n = 90) sets. DL features were extracted from contrast-enhanced MRI images using VGGNet-19. Three feature selection methods and five classification methods were combined for DL signature construction. Subsequently, an mp-MR DL signature fused with multiphase DL signatures of contrast-enhanced images was constructed. Univariate and multivariate logistic regression analyses were used to identify early recurrence risk factors including mp-MR DL signature, microvascular invasion (MVI), and tumor number. A DL nomogram was built by incorporating deep features and significant clinical variables to achieve early recurrence prediction. RESULTS: MVI (p = 0.039), tumor number (p = 0.001), and mp-MR DL signature (p < 0.001) were independent risk factors for early recurrence. The DL nomogram outperformed the clinical nomogram in the training set (AUC: 0.949 vs. 0.751; p < 0.001) and validation set (AUC: 0.909 vs. 0.715; p = 0.002). Excellent DL nomogram calibration was achieved in both training and validation sets. Decision curve analysis confirmed the clinical usefulness of DL nomogram. CONCLUSION: The proposed DL nomogram was superior to the clinical nomogram in predicting early recurrence for HCC patients after hepatectomy. KEY POINTS: • Deep learning signature based on Gd-EOB-DTPA MRI was the predominant independent predictor of early recurrence for hepatocellular carcinoma (HCC) after hepatectomy. • Deep learning nomogram based on clinical factors and Gd-EOB-DTPA MRI features is promising for predicting early recurrence of HCC. • Deep learning nomogram outperformed the conventional clinical nomogram in predicting early recurrence.


Subject(s)
Carcinoma, Hepatocellular , Deep Learning , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/surgery , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/surgery , Liver Neoplasms/blood supply , Hepatectomy , Nomograms , Contrast Media , Gadolinium DTPA , Magnetic Resonance Imaging/methods , Retrospective Studies
8.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 45(5): 721-729, 2023 Oct.
Article in Zh | MEDLINE | ID: mdl-37927012

ABSTRACT

Objective To investigate the expression level of serine/threonine phosphoprotein phosphatase 4C(PPP4C)in gastric cancer,and analyze its relationship with prognosis and the underlying regulatory mechanism.Methods The clinical data of 104 gastric cancer patients admitted to the First Affiliated Hospital of Bengbu Medical College between January 2012 and August 2016 were collected.Immunohistochemical staining was employed to determine the expression levels of PPP4C and Ki-67 in the gastric cancer tissue.The gastric cancer cell lines BGC823 and HGC27 were cultured and transfected with the vector for PPP4C knockdown,the vector for PPP4C overexpression,and the lentiviral vector(control),respectively.The effects of PPP4C on the cell cycle and proliferation were analyzed and the possible regulatory mechanisms were explored.Results PPP4C was highly expressed in gastric cancer(P<0.001),and its expression promoted malignant progression of the tumor(all P<0.01).Univariate and Cox multivariate analysis clarified that high expression of PPP4C was an independent risk factor affecting the 5-year survival rate of gastric cancer patients(P=0.003).Gene ontology and Kyoto encyclopedia of genes and genomes enrichment analysis suggested that PPP4C may be involved in the cell cycle.The correlation analysis showed that the expression of PPP4C was positively correlated with that of Ki-67 in gastric cancer(P<0.001).The up-regulation of PPP4C expression increased the proportion of tumor cells in the S phase,alleviated the G2/M phase arrest,and promoted the proliferation of gastric cancer cells and the expression of cyclin D1 and cyclin-dependent kinase 6(CDK6)(all P<0.05).The down-regulation of PPP4C decreased the proportion of gastric cancer cells in the S phase,promoted G2/M phase arrest,and inhibited cell proliferation and the expression of cyclin D1,CDK6,and p53(all P<0.05).p53 inhibitors promoted the proliferation of BGC823 and HGC27 cells in the PPP4C knockdown group(P<0.001,P<0.001),while p53 activators inhibited the proliferation of BGC823 and HGC27 cells in the PPP4C overexpression group(P<0.001,P=0.002).Conclusions PPP4C is highly expressed in gastric cancer and affects the prognosis of the patients.It may increase the proportion of gastric cancer cells in the S phase and alleviate the G2/M phase arrest by inhibiting p53 signaling,thereby promoting cell proliferation.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Tumor Suppressor Protein p53 , Phosphoproteins/metabolism , Ki-67 Antigen , Cell Line, Tumor , Prognosis , Cell Proliferation , Phosphoprotein Phosphatases/metabolism , Threonine , Serine
9.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 54(6): 1167-1175, 2023 Nov 20.
Article in Zh | MEDLINE | ID: mdl-38162070

ABSTRACT

Objective: The study was conducted to investigate the expression of protein-L-isoaspartate (D-aspartate) O-methyltransferase (PCMT1) in gastric cancer and its effect on the prognosis, and to analyze its potential mechanism. Methods: UALCAN, a cancer data analysis platform, was used to conduct online analysis of the expression of PCMT1 in gastric cancer tissues. Through the Database for Annotation, Visualization and Integrated Discovery (DAVID), Gene Ontology (GO) annotation and signaling pathway enrichment by Kyoto Encyclopedia of Genes and Genomes (KEGG) were performed to analyze the possible functions and signaling pathways. A total of 120 patients who underwent radical gastrectomy for gastric cancer between January 2014 and December 2017 in our hospital were enrolled for the study. Immunohistochemical staining was performed to determine the expression of PCMT1 and Ki67 in gastric cancer tissues. Cox regression, Kaplan-Meier curve, and receiver operating characteristic (ROC) curves were used for prognostic analysis of 5-year survival in gastric cancer patients after surgery. Lentivirus was used to construct PCMT1-interfering or PCMT1-overexpressing vectors, which were then used to transfect human gastric cancer cell lines of MGC-803 and HGC-27 cells. The interfering empty vector (sh-NC) group, the interfering PCMT1 vector (sh-PCMT1) group, the overexpressing empty vector (LV-Vec) group, and the overexpressing PCMT1 vector (LV-PCMT1) group were set up. Western blot was performed to determine the protein expression levels of PCMT1, CyclinB1, and CDC20. CCK-8 assay was performed to measure the proliferation of gastric cancer cells. Flow cytometry was performed to determine the cell cycle. MGC-803 cells were injected in four groups of nude mice to construct a subcutaneous xenograft tumor model, with three nude mice in each group. The body mass of the nude mice was measured. The nude mice were sacrificed after 14 days and the tumor volume was monitored. The expression levels of CyclinB1 and CDC20 proteins in the tumor tissues were determined by Western blot assay. Results: Analysis with UALCAN showed that PCMT1 was highly expressed in gastric cancer tissues. Moreover, elevated expression was found in gastric tumor tissues of different pathological stages and grades and those with lymph node metastasis (P<0.05). GO and KEGG enrichment analyses showed that PCMT1 was mainly involved in the signal regulation of mitosis, spindle assembly checkpoints, and cell cycle. The immunohistochemical results showed that PCMT1 and Ki67 were highly expressed in gastric cancer tissues and that they were positively correlated with each other (P<0.05). Cox multivariate analysis showed that high PCMT1 expression (hazard ratio [HR]=2.921, 95% confidence interval [CI]:1.628-5.239) was one of the independent risk factors affecting the 5-year survival rate of gastric cancer patients after surgery. Kaplan-Meier curve showed that patients with high PCMT1 expression had a lower 5-year survival after surgery (16.7%, HR=4.651, 95% CI: 2.846-7.601) than patients with low PCMT1 expression (70.0%, HR=0.215, 95% CI: 0.132-0.351) did. The ROC curve showed that PCMT1 had an area under the curve (AUC) of 0.764 (95% CI: 0.674-0.854) for predicting 5-year patient survival after surgery. Western blot results showed that lentiviral interference or overexpression of PCMT1 cell lines was successfully constructed. The results of CCK-8 showed that the proliferative ability of MGC-803 and HGC-27 cells was weakened with the downregulation of PCMT1, and the overexpression of PCMT1 promoted cell proliferation (P<0.05). With the interference of PCMT1, the expression of CDC20 protein was decreased, the expression of CyclinB1 protein was increased, and the cell cycle was arrested in the G2/M phase. In contrast, the overexpression of PCMT1 led to the opposite trends (P<0.05). In the sh-PCMT1 group, the tumor volume and mass were decreased and the expression of CDC20 protein was decreased and the expression of CyclinB1 protein was increased in the tumor tissues of the nude mice (P<0.05, compared with those of the sh-NC group. In contrast, the LV-PCMT1 group showed the opposite trends (P<0.05, compared with those of the LV-Vec group). Conclusion: The high expression of PCMT1 in gastric cancer tissues is associated with poor prognosis in patients and may affect tumor cell malignant proliferation via regulating spindle checkpoints in the process of mitosis.


Subject(s)
Stomach Neoplasms , Animals , Mice , Humans , Prognosis , Stomach Neoplasms/pathology , Mice, Nude , M Phase Cell Cycle Checkpoints , Ki-67 Antigen , Sincalide/genetics , Cell Cycle Proteins/genetics , Cell Proliferation , Cell Line, Tumor , Protein D-Aspartate-L-Isoaspartate Methyltransferase/genetics
10.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 54(1): 114-121, 2023 Jan.
Article in Zh | MEDLINE | ID: mdl-36647653

ABSTRACT

Objective: To investigate the prognostic value of the expression of myeloid leukemia factor 1-interacting protein (MLF1IP) in gastric cancer tissue and its regulatory role in tumor progression. Methods: Gene Expression Omnibus (GEO) database was used to analyze the expression level of MLF1IP in tumor tissues of gastric cancer patients. Kaplan-Meier Plotter database was used to analyze the relationship between MLF1IP expression level and patient prognosis. We conducted a retrospective analysis of 108 gastric cancer patients who had undergone radical surgery at our hospital between January 2015 and December 2015. The expression of MLF1IP in gastric cancer tissue and adjacent tissues was examined. We analyzed the relationship between MLF1IP and the clinicopathological parameters of gastric cancer patients and its impact on the long-term prognosis of gastric cancer patients. Univariate and multivariate regression analyses were done to identify the risk factors affecting the long-term prognosis of gastric cancer patients. The assessment value of MLF1IP for long-term prognosis of gastric cancer was analyzed with ROC curve. The effects of MLF1IP on the proliferation, migration, and invasion of gastric cancer cells were analyzed in vitro with gastric cancer cell line (MGC803). A xenograft tumor model was established with nude mice to analyze in vivo the effect of MLF1IP on tumor growth. Results: The results of the gastric cancer cohort GSE29272 of GEO database showed that the expression level of MLF1IP in gastric cancer tissues was significantly higher than that in normal tissues ( P<0.05). Analysis with Kaplan-Meier Plotter database indicated that high MLF1IP expression was correlated with poor prognosis in gastric cancer patients. Immunohistochemical analysis showed that the expression level of MLF1IP in gastric cancer tissues was higher than that in adjacent tissues ( P<0.05). Correlation analysis showed that the MLF1IP level in gastric cancer tissue was positively correlated with Ki67 ( r=0.609, P<0.01), peripheral blood carcinoembryonic antigen (CEA) ( r=0.572, P<0.01) and carbohydrate antigen 19-9 (CA19-9) ( r=0.623, P<0.01). Kaplan-Meier (K-M) survival analysis showed that the 5-year survival rate of patients in the MLF1IP high expression group was significantly lower than that in the MLF1IP low expression group ( P<0.01). Cox regression analysis showed that independent risk factors for 5-year survival after radical gastrectomy for gastric cancer included the expression of MLF1IP ( HR=2.508, 95% CI: 1.259-4.999), CEA≥5 µg/L ( HR=2.171, 95% CI: 1.152-4.092), CA19-9≥37 kU/L ( HR=2.401, 95% CI: 1.094-5.269), and T3-T4 stages ( HR=2.779, 95% CI: 1.049-7.358) and N2-N3 stages ( HR=2.072, 95% CI: 1.100-3.904). ROC analysis showed that the sensitivity, specificity, and accuracy of MLF1IP (the cut-off value was 3.00 relative protein expression level) in assessing the 5-year survival rate after radical gastrectomy for gastric cancer was 75.00%, 76.92%, and 76.2%, respectively ( P<0.05). CCK-8, Transwell assay, and scratch assays showed that in vitro knocking down of MLF1 IP gene expression significantly inhibited the proliferation, migration and invasion of gastric cancer cells. Subcutaneous tumor xenograft experiment in nude mice showed that knocking down MLF1 IP gene significantly inhibited tumor growth. Conclusion: Increased expression of MLF1IP in gastric cancer tissue, which may be involved in the malignant activities of proliferation, migration, and invasion of gastric cancer cells, has a certain predictive value for poor prognosis.


Subject(s)
Leukemia, Myeloid , Stomach Neoplasms , Animals , Mice , Humans , Prognosis , Carcinoembryonic Antigen , Stomach Neoplasms/pathology , Mice, Nude , Retrospective Studies , CA-19-9 Antigen
11.
J Cell Mol Med ; 26(1): 216-227, 2022 01.
Article in English | MEDLINE | ID: mdl-34862717

ABSTRACT

Intestinal barrier dysfunction and intestinal inflammation interact in the progression of Crohn's disease (CD). A recent study indicated that Epac-2 protected the intestinal barrier and had anti-inflammatory effects. The present study examined the function of Epac-2 in CD-like colitis. Interleukin-10 gene knockout (Il-10-/- ) mice exhibit significant spontaneous enteritis and were used as the CD model. These mice were treated with Epac-2 agonists (Me-cAMP) or Epac-2 antagonists (HJC-0350) or were fed normally (control), and colitis and intestinal barrier structure and function were compared. A Caco-2 and RAW 264.7 cell co-culture system were used to analyse the effects of Epac-2 on the cross-talk between intestinal epithelial cells and inflammatory cells. Epac-2 activation significantly ameliorated colitis in mice, which was indicated by reductions in the colitis inflammation score, the expression of inflammatory factors and intestinal permeability. Epac-2 activation also decreased Caco-2 cell permeability in an LPS-induced cell co-culture system. Epac-2 activation significantly suppressed nuclear factor (NF)-κB/mitogen-activated protein kinase (MAPK) signalling in vivo and in vitro. Epac-2 may be a therapeutic target for CD based on its anti-inflammatory functions and protective effects on the intestinal barrier.


Subject(s)
Colitis , Interleukin-10 , Animals , Caco-2 Cells , Colitis/chemically induced , Colitis/drug therapy , Colitis/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism
12.
J Transl Med ; 20(1): 50, 2022 01 29.
Article in English | MEDLINE | ID: mdl-35093120

ABSTRACT

Gastric cancer (GC) is the fifth leading cause of cancer-related death worldwide and is accompanied by low diagnosis and survival rates. The molecular mechanism of GC must be elucidated to improve treatment strategies. Recent research has shown that the expression of myelin and lymphocyte (MAL) protein is reduced in a variety of adenocarcinomas and has the function of suppressing tumor growth. However, the mechanism by which MAL regulates the epithelial-mesenchymal transition (EMT) in GC remains unclear. Here, we showed that MAL expression was downregulated in specimens from patients with GC and was negatively correlated with the clinical stage. Gain- and loss-of function assays showed that interference with MAL significantly increased tumor cell proliferation, metastasis, invasion and the EMT. Overexpression of MAL significantly inhibited the malignant behavior of GC cells. Moreover, MAL suppressed the malignant behavior of GC cells by inhibiting STAT3 phosphorylation in vitro and in vivo. Our data indicate that MAL suppresses the malignant behavior of GC cells via the STAT3/EMT axis. This study also provides insights into the pathophysiological process of GC and a reference for diagnosis and treatment.


Subject(s)
Stomach Neoplasms , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Myelin and Lymphocyte-Associated Proteolipid Proteins , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Phosphorylation , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/pathology
13.
Eur Radiol ; 32(2): 771-782, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34347160

ABSTRACT

OBJECTIVES: We aimed to develop and validate a deep convolutional neural network (DCNN) model for preoperative prediction of microvascular invasion (MVI) in hepatocellular carcinoma (HCC) and its clinical outcomes using contrast-enhanced computed tomography (CECT) in a large population of candidates for surgery. METHODS: This retrospective study included 1116 patients with HCC who had undergone preoperative CECT and curative hepatectomy. Radiological (R), DCNN, and combined nomograms were constructed in a training cohort (n = 892) respectively based on clinicoradiological factors, DCNN probabilities, and all factors; the performance of each model was confirmed in a validation cohort (n = 244). Accuracy and the AUC to predict MVI were calculated. Disease-free survival (DFS) and overall survival (OS) after surgery were recorded. RESULTS: The proportion of MVI-positive patients was respectively 38.8% (346/892) and 35.7 % (87/244) in the training and validation cohorts. The AUCs of the R, DCNN, and combined nomograms were respectively 0.809, 0.929, and 0.940 in the training cohorts and 0.837, 0.865, and 0.897 in the validation cohort. The combined nomogram outperformed the R nomogram in the training (p < 0.001) and validation (p = 0.009) cohorts. There was a significant difference in DFS and OS between the R, DCNN, and combined nomogram-predicted groups with and without MVI (p < 0.001). CONCLUSIONS: The combined nomogram based on preoperative CECT performs well for preoperative prediction of MVI and outcome. KEY POINTS: • A combined nomogram based on clinical information, preoperative CECT, and DCNN can predict MVI and clinical outcomes of patients with HCC. • DCNN provides added diagnostic ability to predict MVI. • The AUCs of the combined nomogram are 0.940 and 0.897 in the training and validation cohorts, respectively.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/surgery , Humans , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/surgery , Neoplasm Invasiveness , Neural Networks, Computer , Nomograms , Retrospective Studies
14.
BMC Gastroenterol ; 22(1): 378, 2022 Aug 08.
Article in English | MEDLINE | ID: mdl-35941537

ABSTRACT

BACKGROUND: The stromal antigen 3 (STAG3) gene encodes an adhesion complex subunit that can regulate sister chromatid cohesion during cell division. Chromosome instability caused by STAG3 gene mutation may potentially promote tumor progression, but the effect of STAG3 on hepatocellular carcinoma (HCC) and the related molecular mechanism are not reported in the literature. The mechanism of the occurrence and development of HCC is not adequately understood. Therefore, the biological role of STAG3 in HCC remains to be studied, and whether STAG3 might be a sensitive therapeutic target in HCC remains to be determined. METHODS: The expression and clinical significance of STAG3 in HCC tissues and cell lines were determined by RT-qPCR and immunohistochemistry analyses. The biological functions of STAG3 in HCC were determined through in vitro and in vivo cell function tests. The molecular mechanism of STAG3 in HCC cells was then investigated by western blot assay. RESULTS: The mRNA expression of STAG3 was lower in most HCC cells than in normal cells. Subsequently, an immunohistochemical analysis of STAG3 was performed with 126 samples, and lower STAG3 expression was associated with worse overall survival in HCC patients. Moreover, cytofunctional tests revealed that the lentivirus-mediated overexpression of STAG3 in HCC cells inhibited cell proliferation, migration, and invasion; promoted apoptosis; induced G1/S phase arrest in vitro; and inhibited tumor growth in vivo. Furthermore, studies of the molecular mechanism suggested that the overexpression of STAG3 increased Smad3 expression and decreased CDK4, CDK6, cyclin D1, CXCR4 and RhoA expression. CONCLUSION: STAG3 exhibits anticancer effects against HCC, and these effects involve the Smad3-CDK4/CDK6-cyclin D1 and CXCR4/RhoA pathways. STAG3 is a tumor-suppressor gene that may serve as a potential target for molecular therapy, which provides a new idea for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6 , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Receptors, CXCR4 , Smad3 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/pharmacology , Up-Regulation , rhoA GTP-Binding Protein/genetics
15.
Neoplasma ; 69(1): 16-27, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34818025

ABSTRACT

Hepatocellular carcinoma (HCC) is a major cause of cancer-related deaths worldwide. Emerging evidence has revealed the vital functions of microRNAs (miRNAs) in cancer malignant progressions. miR-375 has been verified to serve as an antioncogene in tumorigenesis and a potential therapeutic target in various types of cancer. In this study, we aimed to determine the role of miR-375 in the regulation of chemoresistance and metastasis of HCC. Differentially expressed miR-375 and NCAPG2 were externally validated using expression data from The Cancer Genome Atlas (TCGA) database. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect the expression levels of miR-375 in HCC tissues and cell lines. miR-375 mimics and NCAPG2-overexpression were transfected into HepG2 and Huh7 cells to establish miR-375 overexpression models. Cell Counting Kit-8, Transwell, and flow cytometry experiments were conducted to monitor cell proliferation, migration, and apoptosis. The targeting relationship between miR-375 and non-SMC condensin II complex subunit G 2 (NCAPG2) was determined by qRT-PCR, western blot, and luciferase reporter gene assay. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were conducted using Gene Set Enrichment Analysis (GSEA). The pathway enrichment analysis was used to predict the potential pathways for further study. miR-375 was significantly downregulated in HCC tissues and cells compared to adjacent tissue and normal hepatocyte cell line respectively while NCAPG2 was upregulated. The targeting relationship was verified by luciferase reporting assay, and miR-375 could target the 3'UTR of NCAPG2 mRNA and effectively suppress NCAPG2 protein expression. Replenishing of miR-375 significantly repressed HCC cell proliferation and migration, and induced cell apoptosis. Overexpression of NCAPG2 recovered those biological abilities in miR-375 overexpressed cells. Collective data suggested that miR-375 served as a tumor suppressor via regulating NCAPG2. Replenishing of miR-375 or knockout of NCAPG2 could be therapeutically exploited for HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Apoptosis/genetics , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Chromosomal Proteins, Non-Histone , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/genetics , MicroRNAs/genetics
16.
J Cell Mol Med ; 24(13): 7341-7352, 2020 07.
Article in English | MEDLINE | ID: mdl-32463157

ABSTRACT

Epsilon toxin (ETX) is a 33-kDa pore-forming toxin produced by type B and D strains of Clostridium perfringens. We previously found that ETX caused haemolysis of human red blood cells, but not of erythrocytes from other species. The cellular and molecular mechanisms of ETX-mediated haemolysis are not well understood. Here, we investigated the effects of ETX on erythrocyte volume and the role of the putative myelin and lymphocyte (MAL) receptors in ETX-mediated haemolysis. We observed that ETX initially decreased erythrocyte size, followed by a gradual increase in volume until lysis. Moreover, ETX triggered phosphatidylserine (PS) exposure and enhanced ceramide abundance in erythrocytes. Cell shrinkage, PS exposure and enhanced ceramide abundance were preceded by increases in intracellular Ca2+ concentration. Interestingly, lentivirus-mediated RNA interference studies in the human erythroleukaemia cell line (HEL) cells confirmed that MAL contributes to ETX-induced cytotoxicity. Additionally, ETX was shown to bind to MAL in vitro. The results of this study recommend that ETX-mediated haemolysis is associated with MAL receptor activation in human erythrocytes. These data imply that interventions affecting local MAL-mediated autocrine and paracrine signalling may prevent ETX-mediated erythrocyte damage.


Subject(s)
Bacterial Toxins/metabolism , Erythrocytes/metabolism , Erythrocytes/microbiology , Myelin and Lymphocyte-Associated Proteolipid Proteins/metabolism , Phosphatidylserines/metabolism , Calcium/metabolism , Cell Death , Cell Line , Ceramides/metabolism , Erythrocyte Membrane/metabolism , Humans , Protein Binding/drug effects , Reactive Oxygen Species/metabolism
17.
BMC Cancer ; 19(1): 470, 2019 May 17.
Article in English | MEDLINE | ID: mdl-31101029

ABSTRACT

BACKGROUND: To explore prognostic value of the pre-treatment primary lesion apparent diffusion coefficient (ADC) in locoregionally advanced nasopharyngeal carcinoma (LA-NPC). METHODS: A total of 843 patients with newly diagnosed LA-NPC were enrolled from January 2011 to April 2014 and divided into two groups based on ADC values: the low-ADC group and high-ADC group. The 3-year local relapse-free survival (LRFS), distant metastasis free survival (DMFS), disease-free survival (DFS) and overall survival (OS) rates between two groups were compared using Kaplan-Meier curve, and Cox regression analyses were performed to test prognostic value of the pretreatment ADC in LA-NPC. RESULTS: The cut-off value of the pretreatment ADC for predicting local relapse was 784.5 × 10- 6 mm2/s (AUC [area under curve] = 0.604; sensitivity = 0.640; specificity = 0.574), thus patients were divided into low-ADC (< 784.5 × 10- 6; n = 473) group and high-ADC (≥784.5 × 10- 6; n = 370) group. The low-ADC group had significantly higher 3-year LRFS rate and DFS rate than the high-ADC group (LRFS: 96.2% vs. 91.4%, P = 0.003; DFS: 81.4% vs. 73.0%, P = 0.0056). Multivariate analysis showed that the pretreatment ADC is an independent prognostic factor for LRFS (HR, 2.04; 95% CI, 1.13-3.66; P = 0.017) and DFS (HR, 1.41; 95% CI, 1.04-1.89; P = 0.024). CONCLUSIONS: The pretreatment ADC of the primary lesion is an independent prognostic factor for LRFS and DFS in LA-NPC patients.


Subject(s)
Diffusion Magnetic Resonance Imaging , Nasopharyngeal Carcinoma/diagnosis , Nasopharyngeal Neoplasms/diagnosis , Adolescent , Adult , Aged , Child , Disease-Free Survival , Female , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Young Adult
18.
Mol Pharm ; 15(7): 2513-2527, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29757659

ABSTRACT

Mesenchymal stem cells (MSCs) play an important role in cutaneous wound healing; however, the functional mechanisms involved in the healing process are poorly understood. A series of studies indicate that keratinocytes that migrate into the wound bed rely on an epithelial-mesenchymal transition (EMT)-like process to initiate re-epithelialization. We therefore examined whether bone marrow-derived MSCs (BMSCs) could affect biological behavior and induce EMT-like characteristics in the human epidermal keratinocytes (HEKs) and in the immortalized human keratinocyte cell line HaCaT cells, and we investigated the signaling pathways of BMSC-mediated phenotypic changes. By assessing the expression of EMT-related markers including E-cadherin, α-SMA, and Snail family transcription factors by ß2-adrenergic receptor (ß2-AR) blockage using ICI-118,551, a ß2-AR selective antagonist, or ß2-AR small interfering RNA (siRNA), we showed an involvement of ß2-AR signaling in the induction of EMT-like alterations in human keratinocytes in vitro. ß2-AR signaling also affected collective and individual cell migration in human keratinocyte cell lines, which was attenuated by administration of ICI-118,551. Treating the cells with BMSC-conditioned media (BMSC-CM) not only recapitulated the effect of isoproterenol (ISO) on cell migration but also induced the expression of ß2-AR and a panel of proteins associated with mesenchymal phenotype in HEKs and HaCaT cells. Similarly, a blockade of the ß2-AR by either ICI-118,551 or ß2-AR siRNAs reversed both responses of the epidermal keratinocyte cell lines relative to BMSC-CM exposure. These results were further verified in our vivo findings and indicated that the exogenous application of MSCs promoted cutaneous wound healing and endowed the keratinocytes surrounding the wound area with an increased migratory phenotype through activation of ß2-AR signaling. Our findings suggest a biochemical mechanism underlying the function of MSCs in wound re-epithelization, which provides a reliable theoretical basis for the wide application of MSCs in the treatment of chronic wounds.


Subject(s)
Bone Marrow Cells/physiology , Cell Movement/physiology , Keratinocytes/physiology , Mesenchymal Stem Cells/physiology , Wound Healing/physiology , Adrenergic beta-2 Receptor Antagonists/pharmacology , Cell Line , Cell Movement/drug effects , Chronic Disease/therapy , Culture Media, Conditioned/pharmacology , Epidermis/physiology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Humans , Mesenchymal Stem Cell Transplantation , Propanolamines/pharmacology , RNA, Small Interfering/metabolism , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Wound Healing/drug effects , Wounds and Injuries/therapy
19.
Mol Cell Biochem ; 408(1-2): 1-13, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26253173

ABSTRACT

The emerging role of stress-related signaling in regulating cancer development and progression has been recognized. However, whether stress serves as a mechanism to promote gastric cancer metastasis is not clear. Here, we show that the ß2-AR agonist, isoprenaline, upregulates expression levels of CD44 and CD44v8-10 in gastric cancer cells. CD44, a cancer stem cell-related marker, is expressed at high levels in gastric cancer tissues, which strongly correlates with the occurrence of epithelial-mesenchymal transition (EMT)-associated phenotypes both in vivo and in vitro. Combined with experimental observations in two human gastric cancer cell lines, we found that ß2-AR signaling can initiate EMT. It led to an increased expression of mesenchymal markers, such as α-SMA, vimentin, and snail at mRNA and protein levels, and conversely a decrease in epithelial markers, E-cadherin and ß-catenin. Isoprenaline stimulation of ß2-AR receptors activates the downstream target STAT3, which functions as a positive regulator and mediated the phenotypic switch toward a mesenchymal cell type in gastric cancer cells. Our data provide a mechanistic understanding of the complex signaling cascades involving stress-related hormones and their effects on EMT. In light of our observations, pharmacological interventions targeting ß2-AR-STAT3 signaling can potentially be used to ameliorate stress-associated influences on gastric cancer development and progression.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Isoproterenol/pharmacology , Stomach Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics
20.
Chin J Cancer ; 33(10): 511-20, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25104280

ABSTRACT

The parapharyngeal space (PPS) is an inverted pyramid-shaped deep space in the head and neck region, and a variety of tumors, such as salivary gland tumors, neurogenic tumors, nasopharyngeal carcinomas with parapharyngeal invasion, and lymphomas, can be found in this space. The differential diagnosis of PPS tumors remains challenging for radiologists. This study aimed to develop and test a modified method for locating PPS tumors on magnetic resonance (MR) images to improve preoperative differential diagnosis. The new protocol divided the PPS into three compartments: a prestyloid compartment, the carotid sheath, and the areas outside the carotid sheath. PPS tumors were located in these compartments according to the displacements of the tensor veli palatini muscle and the styloid process, with or without blood vessel separations and medial pterygoid invasion. This protocol, as well as a more conventional protocol that is based on displacements of the internal carotid artery (ICA), was used to assess MR images captured from a series of 58 PPS tumors. The consequent distributions of PPS tumor locations determined by both methods were compared. Of all 58 tumors, our new method determined that 57 could be assigned to precise PPS compartments. Nearly all (13/14; 93%) tumors that were located in the pre-styloid compartment were salivary gland tumors. All 15 tumors within the carotid sheath were neurogenic tumors. The vast majority (18/20; 90%) of trans-spatial lesions were malignancies. However, according to the ICA-based method, 28 tumors were located in the pre-styloid compartment, and 24 were located in the post-styloid compartment, leaving 6 tumors that were difficult to locate. Lesions located in both the pre-styloid and the post-styloid compartments comprised various types of tumors. Compared with the conventional ICA-based method, our new method can help radiologists to narrow the differential diagnosis of PPS tumors to specific compartments.


Subject(s)
Diagnosis, Differential , Magnetic Resonance Spectroscopy , Neck/diagnostic imaging , Pharynx/diagnostic imaging , Carcinoma , Humans , Lymphoma/diagnosis , Lymphoma/diagnostic imaging , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/diagnosis , Nasopharyngeal Neoplasms/diagnostic imaging , Nervous System Neoplasms/diagnosis , Nervous System Neoplasms/diagnostic imaging , Radiography , Salivary Gland Neoplasms/diagnosis , Salivary Gland Neoplasms/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL