Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Cell Death Dis ; 12(6): 554, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34050127

ABSTRACT

Distant metastasis accompanied by angiogenesis is the main cause of nasopharyngeal carcinoma (NPC)-related death. Nuclear exosomes (nEXOs) are potential tumour biomarkers. High mobility group box 3 (HMGB3), a nuclear protein, is known to be overexpressed in cancers. However, its role in NPC has not been elucidated. Here, we explore for the first time the function of nEXO HMGB3 in tumour angiogenesis involved in NPC metastasis using a series of in vitro experiments with NPC cell lines and clinical specimens and in vivo experiments with tumour xenograft zebrafish angiogenesis model. We found a high expression of HMGB3 in NPC, accompanied by the formation of micronuclei, to be associated with metastasis. Furthermore, the NPC-secreted HMGB3 expression was associated with tumour angiogenesis. Moreover, HMGB3-containing nEXOs, derived from the micronuclei of NPC cells, were ingested by the human umbilical vein endothelial cells (HUVECs), and accelerated angiogenesis in vitro and in vivo. Importantly, western blotting and flow cytometry analysis showed that circulating nEXO HMGB3 positively correlated with NPC metastasis. In summary, nEXO HMGB3 can be a significant biomarker of NPC metastasis and provide a novel basis for anti-angiogenesis therapy in clinical metastasis.


Subject(s)
Exosomes/metabolism , HMGB3 Protein/metabolism , Nasopharyngeal Carcinoma/blood supply , Nasopharyngeal Neoplasms/blood supply , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Disease Models, Animal , Heterografts , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Nude , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Up-Regulation , Zebrafish
2.
Cell Death Dis ; 12(5): 411, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33875643

ABSTRACT

In nasopharyngeal carcinoma (NPC), the treatment of tumor metastasis and recurrence is challenging and is associated with poor clinical efficacy. Vasculogenic mimicry (VM) is a new blood-supply model of malignant tumor that is closely related to tumors' distant metastasis. Our previous study demonstrated that miR-124 could target Foxq1 to inhibit NPC metastasis. Whether Foxq1 affects metastasis through vasculogenic mimicry is worth consideration. In this study, we show that VM formation positively correlates with the expression of Foxq1, and EGFR, and the TNM stage in 114 NPC patient samples. Meanwhile, we show that VM-positive NPC patients have a poor prognosis. Furthermore, using in vitro and vivo approaches, we confirm that Foxq1 has a significant effect on NPC metastasis through promoting VM formation, which could be effectively inhibited by EGFR inhibitors (Nimotuzumab or Erlotinib). Also a synergistic efficacy of anti-EGFR and anti-VEGF drugs has been found in NPC inhibition. Mechanistically, the luciferase reporter gene and CHIP assays show that Foxq1 directly binds to the EGFR promoter region and regulates EGFR transcription. In conclusion, our results show that Foxq1 is regulated by miR-124 and that it promotes NPC metastasis by inducing VM via the EGFR signaling pathway. Overall, these results provide a new theoretical support and a novel target selection for anti-VM therapy in the treatment of nasopharyngeal carcinoma.


Subject(s)
Forkhead Transcription Factors/metabolism , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/metabolism , Drug Delivery Systems , ErbB Receptors/metabolism , Humans , Nasopharyngeal Carcinoma/blood supply , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/blood supply , Nasopharyngeal Neoplasms/pathology , Neoplasm Metastasis , Signal Transduction
3.
Clin Neurol Neurosurg ; 202: 106537, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33571782

ABSTRACT

OBJECTIVE: Perifocal edema of brain tumors is associated with survival and neurological symptoms. Our aim was to analyze associations between perifocal edema and immunohistochemical features including proliferation potential, microvessel density, neoangiogenesis and invasiveness in brain metastasis (BM). METHODS: 35 patients with BM were included into the retrospective study. The tumors were localized supratentorial in 25 lesions (71.4%) and infratentorial in 10 lesions (28.6%). The following immunohistochemical features were calculated on histopathological specimens: microvessel density, proliferation index Ki 67, matrix-metallopeptidase 9 (MMP9) extracellular matrix metalloproteinase inducer (EMMPRIN) and vascular endothelial growth factor (VEGF) expression. Tumor and edema volumes were estimated semiautomatically on magnetic resonance images. RESULTS: There were no correlations between tumor volume and edema volume. Moreover, no correlation was identified between the investigated immunohistochemical features and tumor/edema volume. In the non-small cell lung cancer subgroup, a positive correlation between tumor volume and VEGF expression was observed (r = 0.52, P = 0.02) and edema volume correlated inversely with MMP9 expression (r = -0.53, P = 0.02). CONCLUSION: In BM, no linear associations exist between tumor volumes, edema volumes and immunohistochemical features reflecting proliferation potential, neoangiogenesis, microvessel density and MMP9 expression. However, in the subgroup of non-small cell lung cancer, there might be associations between MMP9 expression and edema volume as well as between tumor volume and angiogenesis.


Subject(s)
Brain Edema/diagnostic imaging , Brain Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Adult , Aged , Aged, 80 and over , Basigin/metabolism , Bile Duct Neoplasms/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Carcinoma/blood supply , Carcinoma/diagnostic imaging , Carcinoma/metabolism , Carcinoma/secondary , Carcinoma, Non-Small-Cell Lung/blood supply , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Transitional Cell/blood supply , Carcinoma, Transitional Cell/diagnostic imaging , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/secondary , Cell Proliferation , Cholangiocarcinoma/blood supply , Cholangiocarcinoma/diagnostic imaging , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/secondary , Colorectal Neoplasms/pathology , Endometrial Neoplasms/pathology , Female , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Magnetic Resonance Imaging , Male , Matrix Metalloproteinase 9/metabolism , Melanoma/blood supply , Melanoma/diagnostic imaging , Melanoma/metabolism , Melanoma/secondary , Microvascular Density , Middle Aged , Nasopharyngeal Carcinoma/blood supply , Nasopharyngeal Carcinoma/diagnostic imaging , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/secondary , Nasopharyngeal Neoplasms/pathology , Neoplasm Invasiveness , Skin Neoplasms/pathology , Stomach Neoplasms/pathology , Tumor Burden , Urologic Neoplasms/pathology
4.
PLoS One ; 14(8): e0221638, 2019.
Article in English | MEDLINE | ID: mdl-31442259

ABSTRACT

OBJECTIVE: To explore the use of Contrast-enhanced Ultrasound (CEUS) in evaluating angiogenesis in a xenograft nasopharyngeal carcinoma (NPC) model in nude mice and the evolution of CEUS parameters according to the growth of NPC. METHODS: Nude mice were divided into three groups according to experiments conducted at various times from tumor implantation (8 mice/group; group A: 4 weeks from implantation; group B:6 weeks from implantation; group C:8 weeks from implantation). CNE-2 cells were transplanted in 24 nude mice and CEUS evaluations of the tumors were performed at 4, 6 or 8 weeks from implantation. CEUS parametric perfusion images and pathological findings were recorded. R version 3.4.4 software was used to analyze the CEUS parameters and pathological findings. RESULTS: One-way anova analysis indicated statistically significant differences among the three groups with the parameters of peak intensity (PI) (p<0.001), area wash in (AWI) (p<0.001), area wash out (AWO) (p<0.001) and tumor volumes (p<0.001).Pearson correlation coefficient analysis indicated that microvessel density (MVD) was correlated with tumor volume (r = 0.644, p = 0.001), PI (r = 0.904, p<0.0001), AWI (r = 0.547, p = 0.008) and AWO (r = 0.744, P<0.0001). Tumor volume was correlated with MVD (r = 0.644, p = 0.001), PI (r = 0.625, p = 0.002), AWI (r = 0.528, p = 0.012) and AWO (r = 0.784, p<0.001). The percentage of necrosis in histological sections was correlated with the percentage of CEUS unperfused area (r = 0.446,p = 0.038). Spearman rank correlation coefficient analysis indicated that vascular endothelial growth factor (VEGF) was correlated with PI (r = 0.462, P = 0.032). Welch t test indicated PI, AWI and AWO parameters were significantly lower than that of kidneys (p<0.001, p = 0.009, p = 0.005). CONCLUSIONS: The CEUS parameters PI, AWI and AWO indirectly reflect the MVD and the tumor volume in our model of subcutaneous transplanted NPC in nude mice, providing precious information on angiogenesis and tumor growth. VEGF may play a role in promoting angiogenesis of NPC.


Subject(s)
Contrast Media/chemistry , Nasopharyngeal Carcinoma/blood supply , Nasopharyngeal Carcinoma/diagnostic imaging , Neovascularization, Pathologic/diagnostic imaging , Ultrasonography , Animals , Cell Line, Tumor , Humans , Kidney/diagnostic imaging , Kidney/pathology , Mice, Inbred BALB C , Mice, Nude , Microvessels/diagnostic imaging , Microvessels/pathology , Nasopharyngeal Carcinoma/pathology , Neoplasm Staging , Perfusion , Tumor Burden
5.
Eur J Radiol ; 108: 222-229, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30396660

ABSTRACT

PURPOSE: This prospective study aimed to investigate the correlation between two perfusion methods: arterial spin labeling (ASL) and dynamic contrast enhanced (DCE) technique in patients with nasopharyngeal carcinoma (NPC) and to optimize ASL protocol. MATERIALS AND METHODS: Forty-five newly diagnosed NPC patients underwent MR examinations with both 3D pseudo-continuous ASL (pCASL) and DCE-MRI sequences. Tumor blood flow (TBF) derived in pCASL with three post-labeling delay (PLD) times (i.e. 1.0 s, 1.5 s, and 2.0 s) and DCE derived parameters including MaxSlop, contrast enhancement ratio (CER), Initial area under the gadolinium curve (IAUGC), Ktrans, Kep and Ve were measured by two independent observers, and their correlation coefficients were investigated using Spearman test. RESULTS: Inter-observer reproducibility (ICC = 0.931-0.998) was observed to be excellent. Positive correlations between mean, maximum and minimum value of TBFs with different PLDs and DCE-MRI parameters (except Ve) were respectively observed (r = 0.308-0.688, P = 0.000-0.040). CONCLUSION: pCASL may be an alternative method for DCE-MRI in assessing the perfusion level in NPC in the future.


Subject(s)
Nasopharyngeal Carcinoma/blood supply , Nasopharyngeal Neoplasms/blood supply , Adult , Aged , Arteries/physiology , Blood Flow Velocity/physiology , Contrast Media , Female , Humans , Magnetic Resonance Angiography/methods , Magnetic Resonance Imaging/methods , Male , Middle Aged , Nasopharyngeal Neoplasms/pathology , Prospective Studies , Reproducibility of Results , Spin Labels , Young Adult
6.
Biomed Pharmacother ; 108: 1-6, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30212707

ABSTRACT

RIPK4 (receptor interacting serine/threonine kinase 4) has been reported to be aberrantly expressed in several cancer types. However, its expression pattern and functions in nasopharyngeal carcinoma (NPC) have never been reported. In this study, we have shown that the expression of RIPK4 was up-regulated in NPC tissues. RIPK4 promoted the growth and anchorage-independent growth of NPC cells, and down-regulation of RIPK4 inhibited the growth of NPC cells both in the plate-based culture and on the soft agar. Moreover, RIPK4 promoted the expression of VEGF in the NPC cells and induced the tube formation of HUVEC, and Axitinib (the inhibitor for VEGF receptor) inhibited the tumorigenesis driven by RIPK4. In the molecular mechanism study, RIPK4 was found to enhance the interaction between IKKα and IKKß, and activated NF-kB signaling. Taken together, our study demonstrated the oncogenic roles of RIPK4 in NPC and suggested that RIPK4 might be a therapeutic target.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Nasopharyngeal Carcinoma/enzymology , Nasopharyngeal Carcinoma/pathology , Aged , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , I-kappa B Kinase/metabolism , Male , Mice, Nude , Middle Aged , NF-kappa B/metabolism , Nasopharyngeal Carcinoma/blood supply , Nasopharyngeal Carcinoma/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tumor Stem Cell Assay , Up-Regulation/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...