Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 230
Filter
1.
BMC Cancer ; 24(1): 532, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38671389

ABSTRACT

BACKGROUND: Aberrant expressions of desmoglein 2 (Dsg2) and desmocollin 2(Dsc2), the two most widely distributed desmosomal cadherins, have been found to play various roles in cancer in a context-dependent manner. Their specific roles on breast cancer (BC) and the potential mechanisms remain unclear. METHODS: The expressions of Dsg2 and Dsc2 in human BC tissues and cell lines were assessed by using bioinformatics analysis, immunohistochemistry and western blotting assays. Wound-healing and Transwell assays were performed to evaluate the cells' migration and invasion abilities. Plate colony-forming and MTT assays were used to examine the cells' capacity of proliferation. Mechanically, Dsg2 and Dsc2 knockdown-induced malignant behaviors were elucidated using western blotting assay as well as three inhibitors including MK2206 for AKT, PD98059 for ERK, and XAV-939 for ß-catenin. RESULTS: We found reduced expressions of Dsg2 and Dsc2 in human BC tissues and cell lines compared to normal counterparts. Furthermore, shRNA-mediated downregulation of Dsg2 and Dsc2 could significantly enhance cell proliferation, migration and invasion in triple-negative MDA-MB-231 and luminal MCF-7 BC cells. Mechanistically, EGFR activity was decreased but downstream AKT and ERK pathways were both activated maybe through other activated protein tyrosine kinases in shDsg2 and shDsc2 MDA-MB-231 cells since protein tyrosine kinases are key drivers of triple-negative BC survival. Additionally, AKT inhibitor treatment displayed much stronger capacity to abolish shDsg2 and shDsc2 induced progression compared to ERK inhibition, which was due to feedback activation of AKT pathway induced by ERK inhibition. In contrast, all of EGFR, AKT and ERK activities were attenuated, whereas ß-catenin was accumulated in shDsg2 and shDsc2 MCF-7 cells. These results indicate that EGFR-targeted therapy is not a good choice for BC patients with low Dsg2 or Dsc2 expression. Comparatively, AKT inhibitors may be more helpful to triple-negative BC patients with low Dsg2 or Dsc2 expression, while therapies targeting ß-catenin can be considered for luminal BC patients with low Dsg2 or Dsc2 expression. CONCLUSION: Our finding demonstrate that single knockdown of Dsg2 or Dsc2 could promote proliferation, motility and invasion in triple-negative MDA-MB-231 and luminal MCF-7 cells. Nevertheless, the underlying mechanisms were cellular context-specific and distinct.


Subject(s)
Cell Movement , Cell Proliferation , Desmocollins , Desmoglein 2 , Triple Negative Breast Neoplasms , Humans , Desmocollins/metabolism , Desmocollins/genetics , Desmoglein 2/metabolism , Desmoglein 2/genetics , Female , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Cell Line, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Neoplasm Invasiveness , Gene Expression Regulation, Neoplastic , beta Catenin/metabolism , Signal Transduction
2.
Histopathology ; 83(4): 631-646, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37356975

ABSTRACT

AIMS: Gastric adenocarcinoma with enteroblastic differentiation (GAED) is a rare subset of alpha-fetoprotein (AFP)-producing carcinomas with poor prognosis. However, the molecular features associated with the malignant potential of GEAD remain partially elucidated. METHODS AND RESULTS: In this study, the relationship between clinicopathological parameters and aggressive biological behaviour was analysed in 37 patients with GAED. The results showed that GAED tended to infiltrate the deep layer of the gastric wall and possessed more frequent vascular invasion than conventional gastric adenocarcinoma (CGA) (P < 0.001). All distant metastases were observed in the GAED group, not the CGA group (P < 0.001). High HER2 expression was found in nearly 24.32% of the informative cases, and none showed EBV-encoded RNA positivity or deficient mismatch repair. The most frequently mutated gene in GAED was p53. Programmed cell death-ligand 1 (PD-L1) immunostaining revealed 13 patients with a combined positive score (CPS) ≥ 5 (65%, 13 of 20). Thus, based on these molecular markers (immunostaining, in situ hybridisation and mutation analysis), GAED may be classified as a unique subgroup of the chromosomal instability subtype with HER2+ /EBV- /MSS/TP53+ /PD-L1+ . Next-generation sequencing analyses showed that mutations in the TOPI, ELOA and NOTCH3 genes were found only in GAED, and abnormally expressed genes in GAED were significantly enriched in hepatocellular carcinoma-, gland development-, and gastric cancer-related pathways. CONCLUSION: The HER2+ /EBV- /MSS/TP53+ /PD-L1+ profile and hepatocellular carcinoma-related pathways may be significant in the malignant potential of GAED. In addition to anti-HER2 therapy, immune check-point inhibitors may be an effective treatment option for patients with GAED.


Subject(s)
Adenocarcinoma , Carcinoma, Hepatocellular , Liver Neoplasms , Stomach Neoplasms , Humans , Biomarkers, Tumor/analysis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , B7-H1 Antigen , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Liver Neoplasms/genetics , Cell Differentiation/genetics
3.
Cancer Cell Int ; 22(1): 57, 2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35109839

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) often presents with satellite nodules, rendering current curative treatments ineffective in many patients. The heterogeneity of HCC is a major challenge in personalized medicine. The emergence of spatial transcriptomics (ST) provides a powerful strategy for delineating the complex molecular landscapes of tumours. METHODS: In this study, the heterogeneity of tissue-wide gene expression in tumour and adjacent nonneoplastic tissues using ST technology were investigated. The transcriptomes of nearly 10,820 tissue regions and identified the main gene expression clusters and their specific marker genes (differentially expressed genes, DEGs) in patients were analysed. The DEGs were analysed from two perspectives. First, two distinct gene profiles were identified to be associated with satellite nodules and conducted a more comprehensive analysis of both gene profiles. Their clinical relevance in human HCC was validated with Kaplan-Meier (KM) Plotter. Second, DEGs were screened with The Cancer Genome Atlas (TCGA) database to divide the HCC cohort into high- and low-risk groups according to Cox analysis. HCC patients from the International Cancer Genome Consortium (ICGC) cohort were used for validation. KM analysis was used to compare the overall survival (OS) between the high- and low-risk groups. Univariate and multivariate Cox analyses were applied to determine the independent predictors for OS. RESULTS: Novel markers for the prediction of satellite nodules were identified and a tumour clusters-specific marker gene signature model (6 genes) for HCC prognosis was constructed. CONCLUSION: The establishment of marker gene profiles may be an important step towards an unbiased view of HCC, and the 6-gene signature can be used for prognostic prediction in HCC. This analysis will help us to clarify one of the possible sources of HCC heterogeneity and uncover pathogenic mechanisms and novel antitumour drug targets.

4.
J Gastroenterol Hepatol ; 37(4): 714-726, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35062042

ABSTRACT

BACKGROUND AND AIM: Vasculogenic mimicry (VM) is a unique blood supply pattern in malignant tumors that is closely associated with metastasis and poor prognosis. The Hippo signaling effector TAZ is upregulated in several cancers, promoting cancer proliferation and metastasis. This study aimed to identify the function of TAZ and its regulatory mechanism in promoting VM in gastric cancer (GC). METHODS: The expression of TAZ and TEAD4 and their correlations with overall survival and VM-related markers were analyzed in 228 cases of GC. The regulatory mechanism of TAZ and its interaction with TEAD4 in epithelial-mesenchymal transition (EMT) and VM were investigated in vitro and in vivo. RESULTS: TAZ was highly expressed in GC samples and was associated with shorter patient survival time. TAZ expression was positively correlated with TEAD4 and VM in patients with GC. TAZ enhanced the migration and invasion capacity of GC cells through EMT in vitro and upregulated the expression of VM-associated proteins, including VE-cadherin, MMP2, and MMP9, thus promoting VM formation. Overexpression of TAZ accelerated the growth of subcutaneous xenograft and promoted VM formation in vivo. Co-immunoprecipitation assays showed that TAZ can directly bind to TEAD4, and in vitro experiments showed that this binding mediates the function of TAZ in regulating EMT and VM formation in GC. CONCLUSIONS: TAZ promotes GC metastasis and VM by upregulating TEAD4 expression. Our findings expand the role of TAZ in VM and provide new theoretical support for the use of antiangiogenic therapy in the treatment of advanced GC.


Subject(s)
Stomach Neoplasms , TEA Domain Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Cell Line, Tumor , DNA-Binding Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Stomach Neoplasms/pathology , TEA Domain Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/genetics , Up-Regulation
5.
Eur J Med Res ; 26(1): 137, 2021 Dec 04.
Article in English | MEDLINE | ID: mdl-34863306

ABSTRACT

BACKGROUND: Adrenal tuberculosis is difficult to diagnose due to non-specific symptom. Unexpected death due to adrenal insufficiency after trauma surgery is rare. CASE PRESENTATION: A 45-year-old man, who was admitted to hospital because of trauma to the right hand, died unexpectedly on the 13th day after replantation of amputated fingers. He was diagnosed with brain edema and diluted hyponatremia. Autopsy and histopathologic examination revealed severe brain edema combined with cerebellar tonsillar hernia, extensive destruction of adrenal gland caused by bilateral adrenal tuberculosis and right lung invasive pulmonary tuberculosis. CONCLUSIONS: Trauma and pulmonary tuberculosis complicated with adrenal tuberculosis induced the adrenal crisis, which eventually lead to severe cerebral edema and hernia, and finally death from respiratory and circulatory failure. This autopsy and histopathologic examination suggested a possible pathophysiologic mechanism of sudden death due to diluted hyponatremia after trauma surgery.


Subject(s)
Addison Disease/diagnosis , Adrenal Glands/pathology , Death, Sudden/pathology , Tuberculosis/diagnosis , Addison Disease/complications , Autopsy , Bacterial Infections/complications , Bacterial Infections/diagnosis , Brain Edema/complications , Brain Edema/diagnosis , Death, Sudden/etiology , Fatal Outcome , Humans , Hyponatremia/complications , Hyponatremia/diagnosis , Male , Middle Aged , Tuberculosis/complications
6.
BMC Cancer ; 21(1): 1305, 2021 Dec 07.
Article in English | MEDLINE | ID: mdl-34876062

ABSTRACT

BACKGROUND: The expression and activation of eukaryotic translation initiation factor 4E (eIF4E) is associated with cell transformation and tumor initiation, but the functional role and the mechanism whereby it drives immune cell infiltration in breast cancer (BRCA) remain uncertain. METHODS: Oncomine, Timer and UALCAN were used to analyze the expression of eIF4E in various cancers. PrognoScan, Kaplan-Meier plotter, and GEPIA were utilized to analyze the prognostic value of eIF4E in select cancers. In vitro cell experiments were used to verify the role of eIF4E in promoting the progression of BRCA. ImmuCellAI and TIMER database were used to explore the relationship between eIF4E and tumor infiltrating immune cells. The expression of a macrophage marker (CD68+) and an M2-type marker (CD163+) was evaluated using immunohistochemistry in 50 invasive BRCA samples on tissue microarrays. The Human Protein Atlas (HPA) database was used to show the expression of eIF4E and related immune markers. LinkedOmics and NetworkAnalyst were used to build the signaling network. RESULTS: Through multiple dataset mining, we found that the expression of eIF4E in BRCA was higher than that in normal tissues, and patients with increased eIF4E expression had poorer survival and a higher cumulative recurrence rate in BRCA. At the cellular level, BRCA cell migration and invasion were significantly inhibited after eIF4E expression was inhibited by siRNA. Immune infiltration analysis showed that the eIF4E expression level was significantly associated with the tumor purity and immune infiltration levels of different immune cells in BRCA. The results from immunohistochemical (IHC) staining further proved that the expression of CD68+ and CD163+ were significantly increased and correlated with poor prognosis in BRCA patients (P < 0.05). Finally, interaction network and functional enrichment analysis revealed that eIF4E was mainly involved in tumor-related pathways, including the cell adhesion molecule pathway and the JAK-STAT signaling pathway. CONCLUSIONS: Our study has demonstrated that eIF4E expression has prognostic value for BRCA patients. eIF4E may act as an essential regulator of tumor macrophage infiltration and may participate in macrophage M2 polarization.


Subject(s)
Breast Neoplasms/genetics , Eukaryotic Initiation Factor-4E/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/immunology , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Biomarkers, Tumor/genetics , Breast Neoplasms/immunology , Cell Movement/genetics , Cell Movement/immunology , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Invasiveness/genetics , Prognosis , Receptors, Cell Surface/immunology
7.
Front Oncol ; 11: 662434, 2021.
Article in English | MEDLINE | ID: mdl-34249699

ABSTRACT

PURPOSE: Angiotropism is the process by which cancer cells attach to and migrate along blood vessels to acquire vasculature, disseminate, and metastasize. However, the molecular basis for such vessel-tumor interactions has not been fully elucidated, partly due to limited experimental models. In this study, we aimed to observe and explore the molecular mechanism underlying angiotropism in melanoma. METHODS: To monitor the interactions of human melanoma cells with the vasculature in vivo, a murine coxenograft model was employed by co-injecting highly and poorly invasive melanoma cells subcutaneously. To identify key pathways and genes involved in the angiotropic phenotype of melanoma, analysis of differentially expressed genes (DEGs) and gene set enrichment analysis (GSEA) were performed. The role of tissue factor pathway inhibitor 2 (TFPI2) in angiotropism was evaluated by immunostaining, adhesion assay, shRNA, and in vivo tumorigenicity. Angiotropism and TFPI2 expression were examined in surgical specimens of melanoma by immunohistochemical staining. Data from The Cancer Genome Atlas (TCGA) were analyzed to explore the expression and prognostic implications of TFPI2 in uveal and cutaneous melanoma. RESULTS: Highly invasive melanoma cells spread along the branches of intratumor blood vessels to the leading edge of invasion in the coxenograft model, resembling angiotropic migration. Mechanisms underlying angiotropism were primarily associated with molecular function regulators, regulation of cell population proliferation, developmental processes, cell differentiation, responses to cytokines and cell motility/locomotion. TFPI2 downregulation weakened the perivascular migration of highly invasive melanoma cells. High levels of TFPI2 were correlated with worse and better survival in uveal and cutaneous melanoma, respectively. CONCLUSION: These results provide a straightforward in vivo model for the observation of angiotropism and suggest that TFPI2 could inhibit the angiotropic phenotype of melanoma.

8.
Cancer Biol Med ; 2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34086429

ABSTRACT

OBJECTIVE: Hypoxia is a significant feature of solid tumors, including pancreatic ductal adenocarcinoma (PDAC). It is associated with tumor invasion, metastasis, and drug resistance. However, the spatial distribution of hypoxia-related heterogeneity in PDAC remains unclear. METHODS: Spatial transcriptomics (STs), a new technique, was used to investigate the ST features of engrafted human PDAC in the ischemic hind limbs of nude mice. Transcriptomes from ST spots in the hypoxic tumor and the control were clustered using differentially-expressed genes. These data were compared to determine the spatial organization of hypoxia-induced heterogeneity in PDAC. Clinical relevance was validated using the Tumor Cancer Genome Atlas and KM-plotter databases. The CMAP website was used to identify molecules that may serve as therapeutic targets for PDAC. RESULTS: ST showed that the tumor cell subgroups decreased to 7 subgroups in the hypoxia group, compared to 9 subgroups in the control group. Different subgroups showed positional characteristics and different gene signatures. Subgroup 6 located at the invasive front showed a higher proliferative ability under hypoxia. Subgroup 6 had active functions including cell proliferation, invasion, and response to stress. Expressions of hypoxia-related genes, LDHA, TPI1, and ENO1, induced changes. CMAP analysis indicated that ADZ-6482, a PI3K inhibitor, was targeted by the invasive subgroup in hypoxic tumors. CONCLUSIONS: This study is the first to describe hypoxic microenvironment-induced spatial transcriptome changes in PDAC, and to identify potential treatment targets for PDAC. These data will provide the basis for further investigations of the prognoses and treatments of hypoxic tumors.

9.
Cancer Biomark ; 31(3): 227-238, 2021.
Article in English | MEDLINE | ID: mdl-33896817

ABSTRACT

BACKGROUND: Runt-related transcription factor 2 (RUNX2) is an important gene that has been implicated in the progression of human cancer. Aberrant expression of RUNX2 predicts gastric cancer (GC) metastasis. However, the molecular mechanism of RUNX2 remains unknown. OBJECTIVE: We hypothesize that RUNX2 promotes GC metastasis by regulating the extracellular matrix component collagen type I alpha 1 (COL1A1). METHODS: The GEPIA database and immunohistochemical staining of 60 GC tissues were used to analyse the correlations between RUNX2 or COL1A1 expression and clinicopathological features, and the Kaplan-Meier method was used to evaluate survival. RT-PCR, western blotting and immunofluorescence were used to detect RUNX2 and COL1A1 expression in GC cells. Migration and invasion assays were performed to assess the influence of RUNX2 and COL1A1 on metastasis. RESULTS: RUNX2 and COL1A1 were highly expressed at both the gene and protein levels in GC, and patients who were positive for RUNX2 and COL1A1 had shorter survival. RUNX2 and COL1A1 expression linearly correlated with each other (r= 0.15, p< 0.01) and with clinical stage and lymph node metastasis (p< 0.05). Overexpressing RUNX2in vitro enhanced COL1A1 expression and promoted GC cell invasion and migration, whereas COL1A1 knockdown inhibited the increase in cell metastatic capacity promoted by RUNX2. In vivo, GC cells overexpressing RUNX2 promoted lung metastasis, and the downregulation of COL1A1 reduced the metastasis promoted by RUNX2. CONCLUSIONS: RUNX2 may promote GC metastasis by regulating COL1A1. RUNX2/COL1A1 can be employed as a novel target for therapy in GC.


Subject(s)
Collagen Type I, alpha 1 Chain/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Stomach Neoplasms/genetics , Disease Progression , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Stomach Neoplasms/pathology , Transfection
10.
Cancer Biol Med ; 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33710816

ABSTRACT

OBJECTIVE: The immunoscore, which is used to quantify immune infiltrates, has greater relative prognostic value than tumor, node, and metastasis (TNM) stage and might serve as a new system for classification of colorectal cancer. However, a comparable immunoscore for predicting lung adenocarcinoma (LUAD) prognosis is currently lacking. METHODS: We analyzed the expression of 18 immune features by immunohistochemistry in 171 specimens. The relationship of immune marker expression and clinicopathologic factors to the overall survival (OS) was analyzed with the Kaplan-Meier method. A nomogram was developed by using the optimal features selected by least absolute shrinkage and selection operator (LASSO) regression in the training cohort (n = 111) and evaluated in the validation cohort (n = 60). RESULTS: The indicators integrated in the nomogram were TNM stage, neuron-specific enolase, carcino-embryonic antigen, CD8center of tumor (CT), CD8invasive margin (IM), FoxP3CT, and CD45ROCT. The calibration curve showed prominent agreement between the observed 2- and 5-year OS and that predicted by the nomogram. To simplify the nomogram, we developed a new immune-serum scoring system (I-SSS) based on the points awarded for each factor in the nomogram. Our I-SSS was able to stratify same-stage patients into different risk subgroups. The combination of I-SSS and TNM stage had better prognostic value than the TNM stage alone. CONCLUSIONS: Our new I-SSS can accurately and individually predict LUAD prognosis and may be used to supplement prognostication based on the TNM stage.

12.
Nat Commun ; 11(1): 2961, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32528084

ABSTRACT

Colonoscopy is commonly used to screen for colorectal cancer (CRC). We develop a deep learning model called CRCNet for optical diagnosis of CRC by training on 464,105 images from 12,179 patients and test its performance on 2263 patients from three independent datasets. At the patient-level, CRCNet achieves an area under the precision-recall curve (AUPRC) of 0.882 (95% CI: 0.828-0.931), 0.874 (0.820-0.926) and 0.867 (0.795-0.923). CRCNet exceeds average endoscopists performance on recall rate across two test sets (91.3% versus 83.8%; two-sided t-test, p < 0.001 and 96.5% versus 90.3%; p = 0.006) and precision for one test set (93.7% versus 83.8%; p = 0.02), while obtains comparable recall rate on one test set and precision on the other two. At the image-level, CRCNet achieves an AUPRC of 0.990 (0.987-0.993), 0.991 (0.987-0.995), and 0.997 (0.995-0.999). Our study warrants further investigation of CRCNet by prospective clinical trials.


Subject(s)
Colorectal Neoplasms/diagnosis , Deep Learning , Machine Learning , Aged , Colonoscopy , Female , Gastroenterology , Humans , Male , Middle Aged , Retrospective Studies
13.
J Cell Mol Med ; 24(13): 7163-7174, 2020 07.
Article in English | MEDLINE | ID: mdl-32469152

ABSTRACT

The up-regulation of EMT regulator Twist1 has been implicated in vasculogenic mimicry (VM) formation in human triple-negative breast cancer (TNBC). Twist1 targets the Claudin15 promoter in hepatocellular carcinoma cells. Claudin family members are related with TNBC. However, the relationship between Claudin15 and VM formation is not clear. In this study, we first found that Claudin15 expression was frequently down-regulated in human TNBC, and Claudin15 down-regulation was significantly associated with VM and Twist1 nuclear expression. Claudin15 down-regulation correlated with shorter survival compared with high levels. Claudin15 silence significantly enhanced cell motility, invasiveness and VM formation in the non-TNBC MCF-7 cells. Conversely, an up-regulation of Claudin15 remarkably reduced TNBC MDA-MB-231 cell migration, invasion and VM formation. We also showed that down-regulation of Claudin15 was Twist1-dependent, and Twist1 repressed Claudin15 promoter activity. Furthermore, GeneChip analyses of mammary glands of Claudin15-deficient mice indicated that Claudin18 and Jun might be downstream factors of Twist1-Claudin15. Our results suggest that Twist1 induced VM through Claudin15 suppression in TNBC, and Twist1 inhibition of Claudin15 might involve Claudin18 and Jun expression.


Subject(s)
Claudins , Gene Expression Regulation, Neoplastic , Nuclear Proteins , Triple Negative Breast Neoplasms , Twist-Related Protein 1 , Animals , Female , Humans , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/metabolism , Claudins/deficiency , Claudins/genetics , Claudins/metabolism , Kaplan-Meier Estimate , Mammary Glands, Animal/pathology , Neoplasm Invasiveness , Nuclear Proteins/metabolism , Phenotype , Transcription, Genetic , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Twist-Related Protein 1/metabolism , Up-Regulation/genetics , Mice
14.
Cell Oncol (Dordr) ; 43(4): 655-667, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32291572

ABSTRACT

PURPOSE: Upregulation of receptor tyrosine kinase EphA2 has been found to be associated with a poor prognosis in many types of cancer and is considered an attractive therapeutic target. As yet, few efforts have been focused on its tumor suppressive activity triggered by its ligand, ephrinA1. Here, we aimed to determine the potential of ephrinA1 as an important player in melanoma metastasis. METHODS: Data from the Cancer Genome Atlas (TCGA) and the Cancer Cell Line Encyclopedia (CCLE) were analyzed to explore the expression and prognostic implications of EphA2 and ephrinA1 in melanoma. Western blotting, shRNA, colony formation and immunofluorescence assays, as well as two in vivo xenograft models (subcutaneous and metastatic) were used to evaluate the role of EphA2 in melanoma progression. Akt inhibition and ephrinA1-Fc were used to confirm the influence of Akt activation and ephrinA1 levels on the EphA2 effects. Immunohistochemistry (IHC) was performed on xenograft and patient melanoma tissues. RESULTS: We found that high levels of ephrinA1, but not EphA2, were negatively correlated with melanoma metastasis. The expression levels of EphA2 and ephrinA1 were not correlated. After EphA2 downregulation, colony forming abilities and lung metastatic growth were reduced in melanoma cell lines with a low ephrinA1 expression, but were increased in melanoma cell lines with a high ephrinA1 expression. EphA2-mediated colony formation in EphA2-high/ephrinA1-low cells was found to be Akt-dependent and to be inhibited by the addition of ephrinA1-Fc. IHC staining of primary melanoma specimens revealed that EphA2-high/ephrinA1-low patients exhibited poorer outcomes than EphA2-high/ephrinA1-high patients. CONCLUSIONS: From our data we conclude that evaluation of ephrinA1 levels may be helpful for the application of EphA2-targeted therapies and for prognostic predictions in melanoma patients.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Receptor, EphA1/metabolism , Receptor, EphA2/metabolism , Animals , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude
15.
J Cancer ; 11(9): 2560-2571, 2020.
Article in English | MEDLINE | ID: mdl-32201526

ABSTRACT

Vasculogenic mimicry (VM) is linked to vascular invasion of human hepatocellular carcinoma (HCC). BMP4, one BMP family member, is upregulated in several cancers. The purpose of this report is to identify the function of BMP4 in the formation of VM in HCC and the mechanism underling this regulation. In our report, BMP4 up-regulation resulted in an increase in migration, invasion and channel-like structure formation as well as induced epithelial-mesenchymal transition (EMT) process and stem cell-associated proteins OCT4 and SOX2 expression in HCC cells. In addition, The VM-associated proteins, including EphA2, VE-cadherin and MMP2, also could be effectively enhanced by the overexpression of BMP4. Furthermore, according to the TCGA database, higher expression of BMP4 is seen in HCC in contrast to normal liver samples. Immunohistochemistry revealed that BMP4 was positively associated with VM formation, age, histological differentiation, HCC stage, and shorter survival duration. These data demonstrated that BMP4 could promote VM network formation in HCC through induction of stemness in EMT and modulating the EphA2/VE-cadherin/MMP2 signaling pathway.

16.
Int J Oncol ; 56(3): 750-760, 2020 03.
Article in English | MEDLINE | ID: mdl-32124963

ABSTRACT

Pulmonary sarcomatoid carcinomas (PSCs) are a rare subtype of non­small­cell lung cancer and are typically biphasic neoplasms. No effective treatment for PSCs is currently available in clinical practice. The expression of the epithelial­mesenchymal transition (EMT) transcription factors, Twist1, Slug and Snail, as well as the EMT phenotype and vasculogenic mimicry (VM) were analysed in 41 PSC and 79 pulmonary squamous carcinoma (PSCC) samples. Compared with the PSCCs, the PSCs exhibited an EMT phenotype and VM, and they also exhibited an increased expression of the Twist1, Slug, Snail and VM markers. Twist1 expression was associated with metastasis and TNM stage. Twist1­positive patients exhibited a poorer prognosis for overall survival (OS) than those with Twist1­negative PSCs. Transforming growth factor ß1 (TGFß1) was used to induce an EMT transition in a PSCC cell line. SK­MES­1 cells treated with TGFß1 exhibited an increased expression of Twist1. The EMT phenotype, VM and increased migratory and invasive abilities were induced following TGFß1 treatment. Importantly, in cells treated with TGFß1, the EMT phenotype was reversed, VM marker expression was decreased, and the migratory and invasive ability of the PSCC cell line was decreased following Twist1 knockdown. Collectively, this study provides a new perspective of Twist1 in the aggressiveness of PSCs. The identification of Twist1 as an independent marker of poor prognoses may lead to the development of novel strategies for improving the treatment of patients with PSC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Nuclear Proteins/metabolism , Twist-Related Protein 1/metabolism , Up-Regulation , Aged , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Analysis
17.
Endocr Pathol ; 31(1): 39-45, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31788765

ABSTRACT

Napsin A is widely used in the diagnosis of lung adenocarcinoma and has also been reported to be positive in cases of thyroid carcinomas. We investigated napsin A levels through immunohistochemistry on whole sections of 210 primary thyroid tumors of various subtypes and another 41 metastatic thyroid carcinomas, and compared these with 125 primary and 25 metastatic lung adenocarcinomas. The results showed that napsin A was expressed in 23.8% thyroid tumors and 30.3% papillary thyroid carcinomas. Most cases showed a focal and weak to moderate expression. In comparison, 80.8% primary lung adenocarcinomas expressed napsin A, with mostly diffused and strong expression. For metastatic carcinomas of thyroid and lung origin, napsin A was detected in 39.0% of thyroid carcinomas in contrast to 88.0% in cases of lung adenocarcinomas. Comparisons of additional markers, TTF-1, CK7, thyroglobulin, and Pax-8 in metastatic carcinomas showed the overlapping expression of immunomarkers of TTF-1 and CK7. Thyroglobulin and Pax-8 were useful for distinguishing between metastatic carcinomas; however, Pax-8 may be a superior marker due to its higher sensitivity. The clinicopathological analysis of papillary thyroid carcinomas showed that the expression of napsin A was positively correlated with lymph node metastasis (p = 0.030). Here, we focused on the expression of napsin A in thyroid tumors and compared it with that in lung adenocarcinomas. The expression of napsin A is common in thyroid tumors and the combined expression of napsin A and TTF-1 in a metastatic thyroid carcinoma is a cause for concern due to chances of misdiagnosis as lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung/diagnosis , Aspartic Acid Endopeptidases/biosynthesis , Biomarkers, Tumor/analysis , Lung Neoplasms/diagnosis , Thyroid Neoplasms/diagnosis , Aspartic Acid Endopeptidases/analysis , Diagnosis, Differential , Humans
18.
World J Gastroenterol ; 25(35): 5283-5299, 2019 Sep 21.
Article in English | MEDLINE | ID: mdl-31558873

ABSTRACT

BACKGROUND: Highly upregulated in liver cancer (HULC) is a long non-coding RNA (lncRNA) which has recently been identified as a key regulator in hepatocellular carcinoma (HCC) progression. However, its role in the secretion of exosomes from HCC cells remains unknown. AIM: To explore the mechanism by which HULC promotes the secretion of exosomes from HCC cells. METHODS: Serum and liver tissue samples were collected from 30 patients with HCC who had not received chemotherapy, radiotherapy, or immunotherapy before surgery. HULC expression in serum exosomes and liver cancer tissues of patients was measured, and compared with the data obtained from healthy controls and tumor adjacent tissues. The effect of HULC upregulation in HCC cell lines and the relationship between HULC and other RNAs were studied using qPCR and dual-luciferase reporter assays. Nanoparticle tracking analysis was performed to detect the quantity of exosomes. RESULTS: HULC expression in serum exosomes of patients with HCC was higher than that in serum exosomes of healthy controls, and HULC levels were higher in liver cancer tissues than in tumor adjacent tissues. The expression of HULC in serum exosomes and liver cancer tissues correlated with the tumor-node-metastasis (TNM) classification, and HULC expression in tissues correlated with that in serum exosomes. Upregulation of HULC promoted HCC cell growth and invasion and repressed apoptosis. Notably, it also facilitated the secretion of exosomes from HCC cells. Moreover, qPCR assays showed that HULC repressed microRNA-372-3p (miR-372-3p) expression. We also identified Rab11a as a downstream target of miR-372-3p. Dual-luciferase reporter assays suggested that miR-372-3p could directly bind both HULC and Rab11a. CONCLUSION: Our findings illustrate the importance of the HULC/miR-372-3p/Rab11a axis in HCC and provide new insights into the molecular mechanism regulating the secretion of exosomes from HCC cells.


Subject(s)
Carcinoma, Hepatocellular/pathology , Exosomes/metabolism , Liver Neoplasms/pathology , MicroRNAs/genetics , RNA, Long Noncoding/metabolism , rab GTP-Binding Proteins/genetics , Adult , Aged , Apoptosis/genetics , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver/pathology , Liver Neoplasms/blood , Liver Neoplasms/genetics , Male , MicroRNAs/metabolism , Middle Aged , Neoplasm Staging , Up-Regulation
19.
Oncol Lett ; 18(3): 3003-3016, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31452779

ABSTRACT

N-myc downstream regulated gene 1 (NDRG1) has been well characterized as a suppressor of metastasis in numerous types of carcinoma. NDRG1 inhibits the metastatic progression of cancer cells via its inhibitory effects on a wide variety of cellular signaling pathways. Vasculogenic mimicry (VM) refers to the unique ability of aggressive tumor cells to mimic the pattern of embryonic vasculogenic networks, and is the main reason for the poor prognosis and failure of antivascular therapy in gastric carcinoma (GC). Tumor cells can mimic the function of endothelial cells to exhibit VM through epithelial-mesenchymal transition (EMT). However, the potential function of NDRG1 in metastatic GC progression in patients has not yet been fully elucidated. To date, data regarding the function of NDRG1 in VM formation in GC have not been reported. The aim of the present study was to elucidate these unknown areas. To this end, 228 samples of human GC were used to identify the protein expression levels of NDRG1, VM-associated proteins and EMT-associated proteins via immunohistochemistry, and their clinical significance was assessed. In addition, the data of 415 patients with GC were collected from The Cancer Genome Atlas database. A functional enrichment analysis concerning NDRG1 was performed using Metascape and the Gene Set Enrichment Analysis (GSEA). In conclusion, the results of the present study indicate that NDRG1 is negatively correlated with poor prognosis through suppression of VM formation in GC. The results of the present study demonstrated that NDRG1 decreases EMT-associated protein expression and that HER2 expression may serve a significant role in this process. The Metascape and GSEA results also indirectly support this conclusion. The present study discusses the status NDRG1 as a prognostic and selective biomarker in GC, as well as current and future NDRG1-targeted therapies.

20.
Cell Death Dis ; 10(3): 200, 2019 02 27.
Article in English | MEDLINE | ID: mdl-30814488

ABSTRACT

Angiogenesis in solid tumors is divided into two modes: endothelium-dependent vessel (EDV) and vasculogenic mimicry (VM). Sphingosine-1-phosphate receptor 1 (S1PR1) plays a vital role on EDV in a variety of human tumors. However, the relationship between S1PR1 and VM is not clear. The aim of this study is to investigate S1PR1 on the regulation of EDV and mimicry formation in breast cancer. Here we show that S1PR1 phosphorylates the complex of VE-cadherin to regulate the switch of EDV and mimicry formation. Suppression of S1PR1 impairs EDV, but contributes to the generation of VM, invasion, and metastasis in vivo and vitro. By inhibiting RhoA activation, the S1PR1/VE-cadherin signaling is blocked. S1PR1 controls VE-cadherin expression and EDV via RhoA activation. Moreover, the low expression of S1PR1 correlates with VM and poor prognosis in breast cancer patient. The results show that S1PR1 regulated RhoA activation to accelerate VE-cadherin phosphorylation (Y731), leading to increased EDV and reduced VM in breast cancer. S1PR1 may provide a new thinking direction for antiangiogenic therapy for patients with breast cancer.


Subject(s)
Antigens, CD/metabolism , Breast Neoplasms/blood supply , Cadherins/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Female , HEK293 Cells , Heterografts , Human Umbilical Vein Endothelial Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphorylation , Transfection , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...