Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
1.
Pathol Res Pract ; 227: 153644, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34634564

ABSTRACT

OBJECTIVE: Angiogenesis produced by tumor microenvironment is play an important role in development of esophageal squamous cell carcinoma (ESCC). As a quantitative index of angiogenesis, literature has emerged contradictory results about the prognostic role of microvessel density (MVD) in ESCC. The aim of the study was to explore the impact of the correlation between MVD and the prognosis of ESCC based the published evidence. METHODS: Pubmed and Web of science database were screened for the relationship of MVD with prognostic feature in ESCC up to March, 2021. 11 relevant articles were used for meta-analysis. The following data were extracted from the literature: author, year, country, the patients number of high/low MVD, tumor-node-metastasis (TNM) classification, clinical stage, lymphoid infiltrates, vessel invasion, invasive depth, differential degree and survival rate. The hazard ratio (HR) and odds ratios (OR) with 95% CI were used to assess the associations between MVD and overall survival (OS). Chi-squared test and I2 statistics were completed to evaluate the heterogeneity in our study. A random-effects model was used when significant heterogeneity existed (I2>50% and p < 0.05). Egger test was used to calculate the publication bias. Subgroup analysis was stratified by antibody, region, sample capacity to explore the source of heterogeneity. RESULTS: 11 studies with 1055 patients were analyzed. Our results suggested that high MVD is an important factor to advanced TNM classification and clinical stage, and the high MVD is positive correlation with the lymph node invasion and vascular invasion(p < 0.05) in ESCC, but irrelevant to poor differential and invasive depth(p > 0.05). The result also indicated that low MVD is a benefit factor to prolong the survival rate (p < 0.05). And the source of the heterogeneity maybe is that the antibody used to detect the MVD was not consistent, patient number was not large enough and the count method on MVD. CONCLUSION: Across multiple studies, high MVD is correlated with clinicopathological criteria of poor prognosis and survival in ESCC. MVD could be the quantitative index to reactive angiogenesis and may play a pivotal role in ESCC development and progression. MVD may represent a valuable addition to current pathologic analysis and help to guide prognosis and treatment.


Subject(s)
Esophageal Neoplasms/blood supply , Esophageal Squamous Cell Carcinoma/blood supply , Microvascular Density , Microvessels/pathology , Neovascularization, Pathologic , Adult , Aged , Aged, 80 and over , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/therapy , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Prognosis , Risk Assessment , Risk Factors
2.
Int J Biol Sci ; 17(14): 3898-3910, 2021.
Article in English | MEDLINE | ID: mdl-34671207

ABSTRACT

Hypoxia and angiogenesis play key roles in the pathogenesis of esophageal squamous cell carcinoma (ESCC), but regulators linking these two pathways to drive tumor progression remain elusive. Here we provide evidence of ADAM9's novel function in ESCC progression. Increasing expression of ADAM9 was correlated with poor clinical outcomes in ESCC patients. Suppression of ADAM9 function diminished ESCC cell migration and in vivo metastasis in ESCC xenograft mouse models. Using cellular fractionation and imaging, we found a fraction of ADAM9 was present in the nucleus and was uniquely associated with gene loci known to be linked to the angiogenesis pathway demonstrated by genome-wide ChIP-seq. Mechanistically, nuclear ADAM9, triggered by hypoxia-induced translocation, functions as a transcriptional repressor by binding to promoters of genes involved in the negative regulation of angiogenesis, and thereby promotes tumor angiogenesis in plasminogen/plasmin pathway. Moreover, ADAM9 suppresses plasminogen activator inhibitor-1 gene transcription by interacting with its transcription factors at the promoter. Our findings uncover a novel regulatory mechanism of ADAM9 as a transcriptional regulator in angiogenesis and highlight ADAM9 as a promising therapeutic target for ESCC treatment.


Subject(s)
ADAM Proteins/physiology , Esophageal Neoplasms/blood supply , Esophageal Squamous Cell Carcinoma/blood supply , Membrane Proteins/physiology , Neovascularization, Pathologic/physiopathology , Transcription Factors/physiology , Animals , Cell Movement , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic/physiology , Humans , Mice , Mice, SCID , Neovascularization, Pathologic/genetics , Plasminogen Activator Inhibitor 1/genetics , Prognosis , Xenograft Model Antitumor Assays
3.
Clin Transl Med ; 11(7): e459, 2021 07.
Article in English | MEDLINE | ID: mdl-34323409

ABSTRACT

Dysregulated expression of S100A7 is found in several cancers and plays an important role in tumor progression; however, its carcinogenic role in esophageal squamous carcinoma (ESCC) is still poorly understood. Here, we identified that the levels of S100A7 were remarkably upregulated in 341 tumor tissues (P < .001) and 274 serum samples (P < .001) of ESCC patients compared with normal control. It was an independent prognostic factor (P = .026). Furthermore, a new diagnostic model for ESCC based on serum S100A7, SCC, and crfra21-1 was established with area under curve (AUC) up to 0.863 (95% CI: 0.802-0.925). Mechanically, we found upregulated S100A7 could promote cell migration and proliferation through intracellular binding to JAB1 and paracrine interaction with RAGE receptors and then activates the downstream signaling pathways. In addition, exocrine S100A7 could promote M2 macrophage infiltration and polarization by up-regulating M2 macrophage associated proteins, and tumor angiogenesis by enhancing the activation of p-ErK and p-FAK pathways. Further animal experiments confirmed the role of S100A7 in promoting M2 macrophage infiltration and angiogenesis in ESCC. In conclusion, these findings highlighted the potential diagnostic and prognostic value of S100A7 in patients with ESCC. Meanwhile, our results reveal that S100A7 promotes tumor progression by activating oncogenic pathways and remodeling tumor microenvironment, which paving the way for the progress of S100A7 as a therapeutic target for cancer treatment.


Subject(s)
Esophageal Neoplasms/diagnosis , Esophageal Squamous Cell Carcinoma/diagnosis , Macrophages/immunology , S100 Calcium Binding Protein A7/metabolism , Animals , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , COP9 Signalosome Complex/metabolism , Cell Proliferation , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/blood supply , Esophageal Squamous Cell Carcinoma/pathology , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neovascularization, Pathologic , Peptide Hydrolases/metabolism , Prognosis , RNA Interference , RNA, Small Interfering/metabolism , Receptor for Advanced Glycation End Products/metabolism , S100 Calcium Binding Protein A7/antagonists & inhibitors , S100 Calcium Binding Protein A7/blood , S100 Calcium Binding Protein A7/genetics , Signal Transduction
4.
Int J Med Sci ; 17(13): 2013-2023, 2020.
Article in English | MEDLINE | ID: mdl-32788880

ABSTRACT

Non-muscle myosin heavy chain 9 (MYH9) is one novel low frequency mutated gene identified in esophageal squamous cell carcinoma (ESCC) using next-generation sequencing. However, its clinical relevance, potential function and mechanisms remain elusive. Methods: Genomic sequencing datas from 104 esophageal squamous cell carcinoma (ESCC) cases were screened a series of low frequency mutant genes. MYH9 was selected to further analyze its clinical significance, function and PCR-array was performed to explore its potential mechanism. Results: MHY9 is a low frequency mutant gene with a mutation frequency of 2.88% in ESCC. Immunohistochemical analysis showed that MYH9 expression was significantly higher in ESCC tumor tissues, and the expression levels were associated with lymph node metastasis of ESCC patients. Moreover, we found that MYH9 knock-down led to inhibition of cell migration and invasion. PCR-array showed MYH9 knockdown led to a significant change of genes expression associated with angiogenesis and epithelial-to-mesenchymal transition (EMT). This observation is further confirmed in TCGA database of LUSC (lung squamous cell carcinoma), CESC (cervical squamous cell carcinomas) and HNSC (head and neck squamous cell carcinoma). Conclusions: Collectively, our study identifies a novel role and mechanism of MYH9, highlights a significance of MYH9 as a metastatic biomarker, and offers potential therapeutic targets for ESCC patients harboring MYH9 mutations.


Subject(s)
Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Myosin Heavy Chains/genetics , Neovascularization, Pathologic/genetics , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition/physiology , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/blood supply , Esophageal Squamous Cell Carcinoma/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Male , Middle Aged , Mutation , Myosin Heavy Chains/metabolism , Neovascularization, Pathologic/pathology , Prognosis
6.
Med Sci Monit ; 26: e920665, 2020 Apr 24.
Article in English | MEDLINE | ID: mdl-32330123

ABSTRACT

BACKGROUND Protocadherin 8 (PCDH8) functions as a tumor-suppressor gene in many types of cancer. This study aimed to investigate the role of PCDH8 in esophageal squamous cell carcinoma (ESCC). MATERIAL AND METHODS Cell proliferation, apoptosis, transwell assay, tube formation assays, and tumor xenograft experiment were performed to explore the role of PCDH8 in the progression of ESCC. RESULTS PCDH8 was found to be downregulated in ESCC cells. Ectopic expression of PCDH8 blocked proliferation, invasion, and migration and induced apoptosis in ESCC cells. Furthermore, vascular endothelial growth factor A (VEGFA) secretion and the AKT signaling pathway were also inhibited when PCDH8 was upregulated. PCDH8 overexpression suppressed epithelial-mesenchymal transition (EMT) and pro-angiogenic activity of ESCC cells. In a mouse model of ESCC xenograft tumors, PCDH8 overexpression remarkably restrained tumor cell growth, with the tumor inhibition rate of 75.2%. PCDH8 was the target of miR-200c and had a negative correlation with miR-200c. CONCLUSIONS PCDH8 exerts a tumor-suppressive effect against ESCC cells. However, further studies are required to elucidate the exact molecular mechanism underlying the antitumor activity of PCDH8 in ESCC.


Subject(s)
Cadherins/biosynthesis , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/blood supply , Esophageal Squamous Cell Carcinoma/metabolism , 3' Untranslated Regions , Animals , Apoptosis/physiology , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Protocadherins , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
7.
Carcinogenesis ; 41(11): 1543-1552, 2020 11 13.
Article in English | MEDLINE | ID: mdl-32055838

ABSTRACT

Early stage of esophageal squamous cell carcinoma (ESCC) is known to be accompanied by angiogenesis and morphological changes of microvessels. Transcription factor Sox2 is amplified in various cancers including ESCC, but the role of Sox2 in the carcinogenesis and angiogenesis has not been determined. Hence, we aimed to investigate the role of Sox2 in the early stage of ESCC. We found that the expression of Sox2 was significantly higher in early-stage ESCC tissues than that in their adjacent normal tissues. We then established Sox2-inducible normal human esophageal squamous cell line (HetSox2) to investigate the role of Sox2 in esophageal carcinogenesis and angiogenesis in vitro. Sox2 overexpression led to increased cell proliferation and spheroid formation. The culture supernatant of Sox2-overexpressing HetSox2 induced migration and sprouting of endothelial cell line HUVEC (human umbilical vein endothelial cell). As for the mechanism, we found that the expression of secreted protein Suprabasin was directly induced by Sox2. Suprabasin enhanced proliferation of normal human esophageal squamous cells when added to the culture. Moreover, Suprabasin enhanced migration and sprouting of HUVEC cells, which were observed with the culture supernatant of Sox2-overexpressing HetSox2. This angiogenic effect of Suprabasin was abolished by inhibiting AKT phosphorylation, which suggested its dependence on AKT signaling. Finally, we showed that Suprabasin expression and the density of microvessels were significantly higher in ESCC tissues with high Sox2 expression. Our study suggested that increased Sox2 expression in esophageal squamous cells induced Suprabasin expression, and as a result initiated the carcinogenesis via increased cell proliferation and angiogenesis.


Subject(s)
Antigens, Differentiation/metabolism , Biomarkers, Tumor/metabolism , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/pathology , SOXB1 Transcription Factors/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Movement , Cell Proliferation , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/blood supply , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Humans , Neovascularization, Pathologic/metabolism , Phosphorylation , Prognosis , SOXB1 Transcription Factors/genetics , Signal Transduction , Tumor Cells, Cultured
8.
Dig Endosc ; 32(1): 65-73, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31220372

ABSTRACT

OBJECTIVES: Accurate diagnosis of invasion depth is important for reliable treatment of esophageal squamous cell carcinoma (ESCC), but it is limited to the muscularis mucosae to slight submucosal invasion (MM/SM1). The diagnostic accuracy of invasion depth is unsatisfactory and remains to be improved. We aimed to investigate the association between the color of the superficial ESCC and invasion depth using linked color imaging (LCI) under light-emitting diode (LED) light sources. METHODS: Lesions diagnosed as superficial ESCC were observed using white light imaging and then by LCI. The color values were calculated using Commission Internationale de l'Eclariage - L*a*b* color space, and the color difference was calculated according to invasion depth. The vascular diameters and vascular angles of the intrapapillary capillary loops were pathologically analyzed. Their correlation with mucosal color was also investigated by LCI. RESULTS: In all, 52 lesions from 48 patients were analyzed. On the basis of invasion depth, the color difference between the normal mucosa and the lesion was larger in the MM/SM1 or deeper group than in the epithelium and the lamina propria mucosa (EP/LPM) group using LCI (P = 0.025). The vascular diameter was positively correlated with the b* color value (correlation coefficient = 0.302, P = 0.033). CONCLUSION: Observation using LCI under LED light sources may improve the endoscopic diagnosis of the invasion depth of superficial ESCC. Further research is needed to validate its usefulness. (UMIN000024615).


Subject(s)
Esophageal Mucosa/diagnostic imaging , Esophageal Mucosa/pathology , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/diagnostic imaging , Esophageal Squamous Cell Carcinoma/pathology , Aged , Aged, 80 and over , Color , Endoscopic Mucosal Resection , Endoscopy, Gastrointestinal , Esophageal Mucosa/blood supply , Esophageal Mucosa/surgery , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/surgery , Esophageal Squamous Cell Carcinoma/blood supply , Esophageal Squamous Cell Carcinoma/surgery , Esophagectomy , Female , Humans , Lymph Node Excision , Male , Middle Aged , Neoplasm Invasiveness , Retrospective Studies
9.
Mol Oncol ; 14(4): 704-720, 2020 04.
Article in English | MEDLINE | ID: mdl-31733011

ABSTRACT

Anti-angiogenic agents combined with chemotherapy is an important strategy for the treatment of solid tumors. However, survival benefit is limited, urging the improvement of combination therapies. We aimed to clarify the effects of vascular endothelial growth factor receptor 2 (VEGFR2) targeting on hemodynamic function and penetration of drugs in esophagogastric adenocarcinoma (EAC). Patient-derived xenograft (PDX) models of EAC were subjected to long-term and short-term treatment with anti-VEGFR2 therapy followed by chemotherapy injection or multi-agent dynamic contrast-enhanced (DCE-) MRI and vascular casting. Long-term anti-VEGFR2-treated tumors showed a relatively lower flow and vessel density resulting in reduced chemotherapy uptake. On the contrary, short-term VEGFR2 targeting resulted in relatively higher flow, rapid vasodilation, and improved chemotherapy delivery. Assessment of the extracellular matrix (ECM) revealed that short-term anti-angiogenic treatment drastically remodels the tumor stroma by inducing nitric oxide synthesis and hyaluronan degradation, thereby dilating the vasculature and improving intratumoral chemotherapy delivery. These previously unrecognized beneficial effects could not be maintained by long-term VEGFR2 inhibition. As the identified mechanisms are targetable, they offer direct options to enhance the treatment efficacy of anti-angiogenic therapy combined with chemotherapy in EAC patients.


Subject(s)
Adenocarcinoma/drug therapy , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Esophageal Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adenocarcinoma/blood supply , Adenocarcinoma/metabolism , Animals , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/metabolism , Female , Humans , Mice, Nude , Stomach Neoplasms/blood supply , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Cancer Control ; 26(1): 1073274819875736, 2019.
Article in English | MEDLINE | ID: mdl-31533461

ABSTRACT

Cyclin E and hepatocyte growth factor (HGF) have been observed as a multifaceted factor in many cancers, and the assessment of microvascular density (MVD) and micro-lymphatic vessel density (MLVD) has been used to quantify tumor angiogenesis and lymphangiogenesis. The aim of this study was to explore the association between expression of cyclin E, HGF, MVD, and MLVD, and clinicopathologic parameters in esophageal squamous cell carcinoma (ESCC). The expression of cyclin E, HGF, MVD, and MLVD were detected using immunohistochemically anticyclin E, HGF, CD34, and lymphatic vessel endothelial hyaluronan receptor 1 in 168 surgically resected ESCC cases and 30 normal esophageal mucosal samples. The expression levels of cyclin E, HGF, MVD, and MLVD were higher compared to controls. High cyclin E and HGF expression was found more frequently in the tumors larger than 5 cm (P < .001), with poorer differentiation (P = .034) and higher tumor node metastasis (TNM) staging (P = .009) compared to their counterparts. Both MVD and MLVD values were found to be higher in the tumors larger than 5 cm (P < .001), with poorer differentiation (P < .001) and higher TNM staging (P < .001) compared to their counterparts. Furthermore, the expression of MVD and MLVD in both the high cyclin E and high HGF expression groups was significantly higher compared to the low cyclin E and HGF expression groups (P < .001). This study demonstrated that high cyclin E and HGF expression is closely correlated with tumor size, tumor differentiation degree, and TNM stage in patients with ESCC. These findings proposed that cyclin E and HGF could serve as novel molecular markers for preoperational evaluation of ESCC.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/metabolism , Cyclin E/biosynthesis , Esophageal Neoplasms/metabolism , Hepatocyte Growth Factor/biosynthesis , Lymphatic Vessels/metabolism , Neovascularization, Pathologic/metabolism , Aged , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/pathology , Female , Humans , Immunohistochemistry , Lymphatic Vessels/pathology , Male , Middle Aged , Neoplasm Staging , Neovascularization, Pathologic/pathology , Prognosis
11.
Mol Cancer ; 18(1): 1, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30609930

ABSTRACT

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is one of the most lethal malignancies. Neovascularization during tumorigenesis supplies oxygen and nutrients to proliferative tumor cells, and serves as a conduit for migration. Targeting oncogenes involved in angiogenesis is needed to treat organ-confined and locally advanced ESCC. Although the phospholipase C epsilon-1 (PLCE1) gene was originally identified as a susceptibility gene for ESCC, how PLCE1 is involved in ESCC is unclear. METHODS: Matrix-assisted laser desorption ionization time-of-flight mass spectrometry were used to measure the methylation status of the PLCE1 promoter region. To validate the underlying mechanism for PLCE1 in constitutive activation of the NF-κB signaling pathway, we performed studies using in vitro and in vivo assays and samples from 368 formalin-fixed esophageal cancer tissues and 215 normal tissues with IHC using tissue microarrays and the Cancer Genome Atlas dataset. RESULTS: We report that hypomethylation-associated up-regulation of PLCE1 expression was correlated with tumor angiogenesis and poor prognosis in ESCC cohorts. PLCE1 can activate NF-κB through phosphoinositide-phospholipase C-ε (PI-PLCε) signaling pathway. Furthermore, PLCE1 can bind p65 and IκBα proteins, promoting IκBα-S32 and p65-S536 phosphorylation. Consequently, phosphorylated IκBα promotes nuclear translocation of p50/p65 and p65, as a transcription factor, can bind vascular endothelial growth factor-C and bcl-2 promoters, enhancing angiogenesis and inhibiting apoptosis in vitro. Moreover, xenograft tumors in nude mice proved that PLCE1 can induce angiogenesis, inhibit apoptosis, and increase tumor aggressiveness via the NF-κB signaling pathway in vivo. CONCLUSIONS: Our findings not only provide evidence that hypomethylation-induced PLCE1 confers angiogenesis and proliferation in ESCC by activating PI-PLCε-NF-κB signaling pathway and VEGF-C/Bcl-2 expression, but also suggest that modulation of PLCE1 by epigenetic modification or a selective inhibitor may be a promising therapeutic approach for the treatment of ESCC.


Subject(s)
Esophageal Neoplasms/blood supply , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/blood supply , Esophageal Squamous Cell Carcinoma/genetics , Phosphoinositide Phospholipase C/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis/physiology , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Epigenesis, Genetic , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Female , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Nude , Middle Aged , NF-kappa B/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphoinositide Phospholipase C/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction , Up-Regulation , Vascular Endothelial Growth Factor A/genetics
12.
Oncol Rep ; 40(3): 1795-1802, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30015941

ABSTRACT

Angiogenesis is crucial for the progression of esophageal squamous cell carcinoma (ESCC). Anti­angiogenesis by targeting important molecules has been considered as one of the most promising and efficient strategy for cancer therapy. Recent studies have demonstrated that the IQ­domain GTPase activating protein 1 (IQGAP1) plays critical roles in tumorigenesis and cancer progression. We previously reported that IQGAP1 is overexpressed in ESCC, and IQGAP1 knockdown can decrease cell proliferation and metastasis ability in vitro and in vivo. However, the effects of IQGAP1 on the angiogenesis of ESCC and its underlying mechanisms remain unknown. In the present study, we found that IQGAP1 overexpression promoted tumor angiogenesis confirmed by human umbilical vascular endothelial cell (HUVEC) tube formation in vitro and chicken embryo chorioallantoic membrane (CAM) assay in vivo. Moreover, IQGAP1 overexpression in ESCC cells increased expression of vascular endothelial growth factor (VEGF) and phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR2). Meanwhile, we found that levels of AKT and ERK phosphorylation were upregulated in IQGAP1­overexpressing cells. Importantly, IQGAP1­knockdown cells showed the opposing results. Furthermore, AKT and ERK inhibitors not only significantly decreased VEGF expression and VEGFR2 phosphorylation in IQGAP1­overexpressing cells, but also abolished the pro­angiogenic effect of IQGAP1 overexpression on angiogenesis in the HUVEC tube formation and chicken embryo CAM assay. Taken together, this evidence confirms that IQGAP1 overexpression promotes tumor angiogenesis via the AKT and ERK­mediated VEGF­VEGFR2 signaling pathway in ESCC, and IQGAP1 may be an attractive therapeutic target for cancer anti­angiogenesis treatment.


Subject(s)
Esophageal Neoplasms/blood supply , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Pathologic , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , ras GTPase-Activating Proteins/metabolism , Apoptosis , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Chorioallantoic Membrane/metabolism , Chorioallantoic Membrane/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Humans , Phosphorylation , Tumor Cells, Cultured
13.
Esophagus ; 15(1): 19-26, 2018 01.
Article in English | MEDLINE | ID: mdl-29892805

ABSTRACT

BACKGROUND: The relationship between thymidine phosphorylase (TP) and angiogenesis at the early stage of esophageal squamous cell carcinoma has been unclear. METHODS: Using 14 samples of normal squamous epithelium, 11 samples of low-grade intraepithelial neoplasia, and 64 samples of superficial esophageal cancer, microvessel density (MVD) was estimated using immunostaining for CD34 and CD105. TP expression was also evaluated in both cancer cells and stromal monocytic cells (SMCs). We then investigated the correlation between MVD and TP expression in both cancer cells and SMCs. RESULTS: On the basis of the above parameters, MVD was significantly higher in cancerous lesions than in normal squamous epithelium. In terms of CD34 and CD105 expression, MVD showed a gradual increase from normal squamous epithelium, to low-grade intraepithelial neoplasia, and then to M1 and M2 cancer, and M3 or deeper cancer. M1 and M2 cancer showed overexpression of TP in both cancer cells and SMCs. There was no significant correlation between TP expression in cancer cells and MVD estimated from CD34 (rS = 0.16, P = 0.21) or CD105 (rS = 0.05, P = 0.68) expression. Significant correlations were found between TP expression in SMCs and CD34-related (rS = 0.46, P < 0.001) and CD105-related (rS = 0.34, P < 0.01) MVD. In M3 or deeper cancers, there were no significant correlations between TP expression in cancer cells or SMCs and venous invasion, lymphatic invasion, and lymph node metastasis. CONCLUSION: TP expression is activated in both cancer cells and stromal monocytic cells at the very early stage of ESCC progression. TP expression in SMCs, rather than in cancer cells, is significantly correlated with angiogenesis.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Esophageal Neoplasms/enzymology , Neovascularization, Pathologic/enzymology , Thymidine Phosphorylase/physiology , Antigens, CD34/metabolism , Carcinoma, Squamous Cell/blood supply , Disease Progression , Endoglin/metabolism , Epithelium/blood supply , Epithelium/enzymology , Esophageal Neoplasms/blood supply , Esophageal Squamous Cell Carcinoma , Esophagus/blood supply , Esophagus/enzymology , Humans , Microvessels/pathology , Precancerous Conditions/enzymology , Stromal Cells/enzymology , Thymidine Phosphorylase/metabolism
14.
Mol Med Rep ; 18(1): 131-138, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29749455

ABSTRACT

Aidi injection, a proprietary Chinese medicine, has been widely used for the treatment of cancer. However, its effects and potential mechanism in esophageal squamous cell carcinoma (ESCC) have not yet been characterized. The aim of the present study was to identify the mechanism underlying the anti­metastatic effects of treatment with Aidi. To test the effects and mechanism, EC9706 and KYSE70 cells were selected for in vitro experiments. In vivo, the anti­metastatic effects of Aidi injection on a nude mouse peritoneal metastasis model were examined, and the mechanisms were assessed with immunohistochemical staining. A cell proliferation assay demonstrated that treatment with more than 3 mg/ml Aidi for 24 or 48 h significantly inhibited the proliferation of EC9706 (P<0.01) and KYSE70 cells (P<0.05, P<0.01). Subsequent experiments assessed cell migration, invasion and vasculogenic mimicry (VM) formation, with 5­fluorouracil serving as a positive control. It was observed that treatment with Aidi inhibited cell migration, invasion and VM formation. Furthermore, it was identified that treatment with Aidi inhibited epithelial­mesenchymal transition (EMT) signaling and the expression of vascular endothelial growth factor A (VEGF­A) in EC9706 and KYSE70 cells, using western blotting. In the in vivo experiments, Aidi injection effectively suppressed tumor metastasis in the mouse tumor model. Additionally, the expression of vimentin and vascular endothelial growth factor was decreased, and the expression of cadherin­1 was increased in the tumor tissue. The present results suggested that treatment with Aidi may inhibit tumor metastasis in ESCC through the inhibition of EMT signaling and angiogenesis.


Subject(s)
Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/drug therapy , Drugs, Chinese Herbal/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
15.
Dis Esophagus ; 30(11): 1-5, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28881911

ABSTRACT

Observation of the microvasculature using narrow band imaging (NBI) with magnifying endoscopy is useful for diagnosing superficial squamous cell carcinoma. Increased vascular density is indicative of cancer, but not many studies have reported differences between cancerous and noncancerous areas based on an objective comparison. We observed specimens of endoscopic submucosal dissection (ESD) using NBI magnification, and determined the vascular density of cancerous and noncancerous areas. A total of 25 lesions of esophageal squamous cell carcinoma that were dissected en bloc by ESD between July 2013 and December 2013 were subjected to NBI magnification. We constructed a device that holds an endoscope and precisely controls the movement along the vertical axis in order to observe submerged specimens by NBI magnification. NBI image files of both cancerous (pathologically determined invasion depth, m1/2) and surrounding noncancerous areas were created and subjected to vascular density assessment by two endoscopists who were blinded to clinical information. The invasion depth was m1/2 in 20, m3/sm1 in four and sm2 in one esophageal cancer lesion. Mean vascular density was significantly increased in cancerous areas (37.6 ± 16.3 vessels/mm2) compared with noncancerous areas (17.6 ± 10.0 vessels/mm2) (P < 0.05). The correlation coefficients between vascular density determined by two endoscopists were 0.86 and 0.81 in cancerous and noncancerous areas, respectively. Receiver operating curve (ROC) analysis revealed that the area under the curve (AUC) of vascular density was 0.895 (95% CI, 0.804-0.986). For this ROC curve, sensitivity was 78.3% and specificity was 87.0% when the cutoff value of vascular density was 26 vessels/mm2. NBI magnification confirmed significant increases in vascular density in cancerous areas compared with noncancerous areas in esophageal squamous cell carcinoma. The rates of agreement between vascular density values determined by two independent operators were high.


Subject(s)
Carcinoma, Squamous Cell/blood supply , Esophageal Neoplasms/blood supply , Esophagoscopy/methods , Esophagus/blood supply , Microvessels/pathology , Narrow Band Imaging/methods , Aged , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Esophagus/pathology , Female , Humans , Image Enhancement/methods , Male , Microvessels/diagnostic imaging , Middle Aged , ROC Curve , Sensitivity and Specificity
16.
Medicine (Baltimore) ; 96(29): e7600, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28723804

ABSTRACT

BACKGROUND: To date, literature has emerged that shows contradictory results about the prognostic role of microvessel density (MVD) in esophageal squamous cell cancer (ESCC). The aim of the study set out to evaluate the correlation between MVD and the prognosis of ESCC. METHODS: Identified publications from various databases were obtained and reviewed. A meta-analysis was performed to evaluate the prognostic role of MVD among ESCC patients. RESULTS: A total of 11 eligible studies containing 891 ESCC cases were included in the meta-analysis. The pooled hazard ratio for overall survival was 2.39 (95% confidence interval 1.92-2.96, P < .001). Heterogeneity among the studies was not significant, and publication bias was not found. Subgroup analyses were also performed on different issues, such as districts, antibodies, and median age. CONCLUSION: High MVD is a prognostic factor among ESCC that indicated worse prognosis in these patients. More studies are needed, and through abundant evidence, the topic could be re-evaluated by then.


Subject(s)
Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/diagnosis , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/diagnosis , Microvessels , Esophageal Squamous Cell Carcinoma , Humans , Prognosis
18.
Hematol Oncol Clin North Am ; 31(3): 499-510, 2017 06.
Article in English | MEDLINE | ID: mdl-28501090

ABSTRACT

Antiangiogenesis therapy is one of only 2 biologically targeted approaches shown to improve overall survival over standard of care in advanced adenocarcinoma of the stomach or gastroesophageal junction (GEJ). Therapeutic targeting of vascular endothelial growth factor receptor 2 improves overall survival in patients with previously treated advanced gastric/GEJ adenocarcinoma. No antiangiogenesis therapy has demonstrated an overall survival benefit in patients with chemo-naïve or resectable esophagogastric cancer or in patients whose tumors arise from the esophagus. Promising ongoing clinical investigations include the combination of antiangiogenesis therapy with immune checkpoint inhibition and anti-human epidermal growth factor receptor 2 therapy.


Subject(s)
Adenocarcinoma/drug therapy , Angiogenesis Inhibitors/therapeutic use , Esophageal Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Stomach Neoplasms/drug therapy , Adenocarcinoma/blood supply , Esophageal Neoplasms/blood supply , Esophagogastric Junction/blood supply , Humans , Stomach Neoplasms/blood supply
19.
Nihon Geka Gakkai Zasshi ; 118(1): 25-30, 2017 Jan.
Article in Japanese | MEDLINE | ID: mdl-30176133

ABSTRACT

Preoperative simulation of vascular anatomy has been widely accepted in order to reduce surgical complications and improve postoperative outcomes. In esophagectomy, preservation of the bronchial artery (BA) was shown to reduce postoperative pulmonary complications. However, some anomalous BA branching patterns have been reported and these can make BA preservation difficult during surgery. Recently, the clinical utility of preoperative three-dimensional computed tomography angiography (3D-CTA) has been reported as a form of preoperative anatomical simulation. Consequently, the BA was safely preserved and efficient lymph node (LN) dissection was achieved. In surgery for gastric cancer, tracing the inner dissectable layer is necessary for LN dissection. Particularly in laparoscopic total gastrectomy with spleen preservation, there is considerable variation in the vascular anatomy of the splenic artery, splenic vein, and short gastric artery. Therefore, preoperative 3D-CTA could improve the safety of this procedure. Recently, the number of dissected LNs has been shown to be increased after introduction of 3D-CTA in laparoscopic surgery for both esophageal and gastric cancer, which showed that preoperative anatomical simulation could achieve more radical LN dissection. As a future perspective, intraoperative navigation systems could become more practical guides for endoscopic surgery for upper gastrointestinal cancer.


Subject(s)
Esophageal Neoplasms/surgery , Stomach Neoplasms/surgery , Angiography , Esophageal Neoplasms/blood supply , Esophagectomy , Esophagoscopy , Gastrectomy , Gastroscopy , Humans , Imaging, Three-Dimensional , Lymph Node Excision , Lymph Nodes/surgery , Stomach Neoplasms/blood supply
20.
Gan To Kagaku Ryoho ; 44(12): 2006-2008, 2017 Nov.
Article in Japanese | MEDLINE | ID: mdl-29394849

ABSTRACT

INTRODUCTION: Chemoradiotherapy(CRT)is an effective treatment method for esophageal cancer. In early stages, it is a standard therapy combined with surgery. However, CRT achieves definitive complete response(CR)in only about 20% of advanced cancer with invasion into adjacent organs. Then, surgery is the only treatment for curative therapy. We report a case of a patient with 3-year survival who underwent lymphadenectomy for residual cancer after CRT for advanced esophageal cancer with invasion into the trachea and right cervical artery. CASE DESCRIPTION: The patient was a 71-year-old woman. Various examinations revealed a cervical esophageal cancer, which had a right cervical lymph node metastasis with invasion into the trachea and right common cervical artery(cT4b[LYM-Tr, RCCA], N1, M0, cStage III C(UICC TNM classification). Induction chemotherapy(DCF; docetaxel[DTX]plus cisplatin[CDDP]plus 5-fluorouracil[5-FU])was initiated, but neither the cancer primary site nor the lymph node metastasis decreased. Then, she received chemoradiotherapy(5-FU plus CDDP and 40.8 Gy). After that, endoscopic and pathological examination showed CR of the primary site, but CT still indicated the presence of a residual lesion in the lymph node. As we diagnosed the residual tumor as being close to the trachea and RCCA, but not infiltrating them, lymphadenectomy was performed, which was possible to preserve the trachea and RCCA. The postoperative histopathological report indicated lymph node metastasis in the right cervical lymph node with a negative radial margin. It has now been about 3 years since her operation, and she is alive and disease-free.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carotid Arteries/pathology , Chemoradiotherapy , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/therapy , Trachea/pathology , Aged , Cisplatin/administration & dosage , Docetaxel , Esophageal Neoplasms/pathology , Female , Fluorouracil/administration & dosage , Humans , Lymph Node Excision , Lymphatic Metastasis , Neoplasm Invasiveness , Taxoids/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...